EP2174131A1 - Criblage, thérapie et diagnostic - Google Patents

Criblage, thérapie et diagnostic

Info

Publication number
EP2174131A1
EP2174131A1 EP08786360A EP08786360A EP2174131A1 EP 2174131 A1 EP2174131 A1 EP 2174131A1 EP 08786360 A EP08786360 A EP 08786360A EP 08786360 A EP08786360 A EP 08786360A EP 2174131 A1 EP2174131 A1 EP 2174131A1
Authority
EP
European Patent Office
Prior art keywords
trem
ligand
receptor
sepsis
binding
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP08786360A
Other languages
German (de)
English (en)
Inventor
Marco Colonna
Julia Klesney-Tait
Paola Panina
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Novo Nordisk AS
Original Assignee
Bioxell SpA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bioxell SpA filed Critical Bioxell SpA
Publication of EP2174131A1 publication Critical patent/EP2174131A1/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • G01N33/5047Cells of the immune system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56966Animal cells
    • G01N33/56972White blood cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/06Gastro-intestinal diseases
    • G01N2800/065Bowel diseases, e.g. Crohn, ulcerative colitis, IBS
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/26Infectious diseases, e.g. generalised sepsis

Definitions

  • the present invention relates to the diagnosis and treatment of inflammatory disorders. It also relates to screening methods for identifying drugs or drug candidates of potential use in treating inflammatory disorders and particularly sepsis and inflammatory bowel disease (IBD).
  • IBD inflammatory bowel disease
  • TREM-1 is a cell-surface molecule that has been identified both on human and murine polymorphonuclear neutrophils and mature monocytes [Bouchon et al, J. Immunol. 164: 4991-4995 (2000)]. It belongs to the immunoglobulin superfamily and activates downstream signalling pathways with the help of an adapter protein called DAP12 [Bouchon et al, supra; Colonna et al, J. Infect. Dis. 187 (Suppl): S297-301 (2003), Colonna Nat. Rev. Immunol. 6:445- 453 (2003); Nathan et al Nat. Med. 7:530-2 (2001 )].
  • TREM-1 Triggering via TREM-1 results in the production of pro-inflammatory cytokines, chemokines, reactive oxygen species, and leads to rapid degranulation of neutrophilic granules, and phagocytosis. It has been shown that blockade of TREM-1 signaling suppresses the development of collagen-induced arthritis (Abstract presented by Y. Murakami at: Innate Immunity: Signaling Mechanisms February 2008, Keystone, Colorado). Furthermore, TREM-1 activation, by dampening LPS-induced IL-12 family cytokines, may impact T cell responses in vivo, thus suggesting an in vivo role of TREM-1 activation not only in innate but also in adaptive immune responses (Dower K, J. Immunol. 2008 180:3520-3534).
  • TREM-1 Since interfering with TREM-1 engagement leads to the simultaneous reduction in production and secretion of a variety of proinflammatory mediators, TREM-1 represents an attractive target for treating chronic inflammatory disorders. Indeed, a role for TREM-1 has been demonstrate in a variety of inflammatory disorders, including acute endotoxemia, Helicobacter pylori infection, hepatic granulomatosis, Salmonella enterica infection, Infectious lung diseases, Marburg and Ebola viruses infections, Acute respiratory distress syndrome (ARDS), inflammatory bowel disease and rheumatoid arthritis, sepsis.
  • acute endotoxemia Helicobacter pylori infection
  • hepatic granulomatosis Salmonella enterica infection
  • Infectious lung diseases Marburg and Ebola viruses infections
  • ARDS Acute respiratory distress syndrome
  • inflammatory bowel disease and rheumatoid arthritis
  • Sepsis constitutes a significant consumption of intensive care resources and remains an ever- present problem in the intensive care unit. It has been estimated that between 400,000 and 500,000 patients are so affected each year in both the USA and Europe. Morbidity and mortality have remained high despite improvements in both supportive and anti-microbial therapies. Mortality rates vary from 40% for uncomplicated sepsis to 80% in those suffering from septic shock and multi-organ dysfunction. The pathogenesis of the conditions is now becoming better understood. Greater understanding of the complex network of immune, inflammatory and haematological mediators may allow the development of rational and novel therapies. Following an infection, innate and cognitive immune responses develop in sequential phases that build-up in specificity and complexity, resulting ultimately in the clearance of infectious agents and restoration of homeostasis.
  • the innate immune response serves as the first line of defence and is initiated upon activation of pattern recognition receptors, such as Toll-like receptors (TLRs) [Aderem et al, Nature 406:782-786 (2000) and Thoma-Uszynski et al, Science 291 :1544- 1549(2001 )].
  • TLRs Toll-like receptors
  • PAMPs pathogen-associated microbial patterns
  • TLRs Activation of the TLRs triggers the release of large quantities of such cytokines as TNF- ⁇ and IL-1 ⁇ , which, in case of such massive infections as sepsis, can precipitate tissue injury and lethal shock [Cohen, et al, Nature 420:885- 91 (2002); Hotchkiss et al, N. Engl. J. Med. 348:138-50 (2003)].
  • TREM-1 myeloid cells-1
  • TREM-1 is a cell-surface molecule that has been identified both on human and murine polymorphonuclear neutrophils and mature monocytes [Bouchon et al, J. Immunol. 164: 4991-4995 (2000)]. It belongs to the immunoglobulin superfamily and activates downstream signalling pathways with the help of an adapter protein called DAP12 [Bouchon et al, supra; Colonna et al, J. Infect. Dis. 187 (Suppl): S297-301 (2003), Colonna Nat. Rev. Immunol. 6:445- 453 (2003); Nathan et al Nat. Med. 7:530-2 (2001 )].
  • TREM-1 was greatly up-regulated on neutrophils and monocytes in the presence of bacteria such as Pseudomonas aeruginosa or Staphylococcus aureus, both in cell culture and in tissue samples from patients with infection [Bouchon et al, Nature 410:1103-1107 (2001 )].
  • TREM-1 was not up-regulated in samples from patients with non-infectious inflammatory diseases such as psoriasis, ulcerative colitis or vasculitis caused by immune complexes [Bouchon et al, Nature 410:1103-1107 (2001 )].
  • TREM-1 when TREM-1 is bound to its ligand, there is a synergistic effect of LPS and an amplified synthesis of the pro-inflammatory cytokines TNF- ⁇ and GM-CSF, together with an inhibition of IL-10 production [Bleharski et al. J. Immunol. 170:3812-3818 (2003)].
  • blockade of TREM-1 signalling protected the animals from death, further highlighting the crucial role of this molecule [Colonna et al, J. Infect. Dis. 187 (Suppl):S297-301 (2003), Bouchon et al, Nature 410:1103-1107 (2001 )].
  • TREM-1 plays a critical role in the inflammatory response to infection [Bouchon et al. J. Immunol. 164:4991-4995 (2000)]. Expression of TREM-1 is increased on myeloid cells in response to both bacterial and fungal infections in humans. Similarly, in mice the induction of shock by lipopolysaccharide (LPS) is associated with increased expression of TREM- 1. Further, treatment of mice with a soluble TREM-1 /Ig fusion protein, as a 'decoy' receptor, protects mice from death due to LPS or E.coli.
  • LPS lipopolysaccharide
  • SIRS systemic inflammatory response syndrome
  • Sepsis is further stratified into severe sepsis when there is evidence of organ hypoperfusion, made evident by signs of organ dysfunction such as hypoxaemia, oliguria, lactic acidosis or altered cerebral function.
  • Septic shock is severe sepsis complicated by hypotension defined as systolic blood pressure less than 90 mmHg despite adequate fluid resuscitation.
  • Sepsis and SIRS may be complicated by the failure of two or more organs, termed multiple organ failure (MOF), due to disordered organ perfusion and oxygenation.
  • MOF multiple organ failure
  • a systemic inflammatory response may occur in severe inflammatory conditions such as pancreatitis and burns.
  • gram-negative bacteria are implicated in 50 to 60% of sepsis with gram-positive bacteria accounting for a further 35 to 40% of cases. The remainder of cases are due to the less common causes of fungi, viruses and protozoa.
  • TREM-1 receptor a ligand for the TREM-1 receptor and have confirmed that it is expressed upon neutrophils and monocytes from septic patients.
  • a screening method comprising providing a TREM-1 ligand or a derivative thereof and determining whether or not a test compound affects:
  • the method is preferably used for screening for compounds that are useful in the treatment of TREM-1 related inflammatory disorders, particularly sepsis and Inflammatory Bowel Disease (IBD).
  • TREM-1 related inflammatory disorders particularly sepsis and Inflammatory Bowel Disease (IBD).
  • the method can be used for screening for drugs/drug candidates.
  • the method desirably comprises the step of determining whether or not a test compound blocks or reduces the binding of the TREM-1 ligand / derivative thereof to the TREM-1 receptor / derivative or whether or not it blocks or reduces an activity that is mediated by said binding.
  • the compound is concluded to be potentially useful in the treatment of TREM-1 related inflammatory disorders, particularly sepsis and Inflammatory Bowel Disease (IBD) .
  • the method is used for screening for compounds useful in the treatment of sepsis mediated by a pathogen.
  • pathogen is used herein to describe any infectious organism that can be detrimental to the health of a human or non-human animal host.
  • TREM-1 ligand is a useful marker of pathogen-mediated sepsis and can be used to distinguish this from SIRS conditions where there is no pathogenic involvement.
  • sepsis may be due to a microbial infection.
  • the infection may for example be bacterial, fungal, protozoal or viral.
  • the method uses cells that express a TREM-1 receptor or at least a ligand-binding part of a TREM-1 receptor.
  • the intracellular part of a TREM-1 receptor may be replaced with a heterologous moiety that is normally not associated with the TREM-1 receptor. This is useful in certain reporter based screening systems.
  • the cytoplasmic region of CD3 ⁇ may be used, as discussed later in Example 11.
  • the cells may be those that express the receptor naturally. Thus they may be neutrophils or monocytes. Such cells can be obtained from patients with sepsis. Alternatively, the cells need not be neutrophils or monocytes, but may be other cells that that do not normally express the TREM- 1 receptor, but have been modified to express the TREM-1 receptor or a TREM-1 ligand binding part thereof. Modification may be performed by techniques known in the art. For example, the cells may be transfected with a vector encoding the TREM-1 receptor or at least the ligand binding part of this receptor and a suitable promoter (e.g. an inducible or constitutive promoter).
  • a suitable promoter e.g. an inducible or constitutive promoter
  • the cells may be heterologous cells, relative to cells in which the receptor is normally expressed.
  • TREM-1 receptor/ ligand binding part thereof it is not however even essential for the TREM-1 receptor/ ligand binding part thereof to be associated with a cell. Soluble forms may be used. Multimeric forms may even be used.
  • a tetramer comprising four soluble forms linked to a streptavidin scaffold may be used, as described in greater detail later on herein.
  • a soluble form comprising the TREM-1 receptor extracellular domain fused to IgG contant regions may be used.
  • TREM-1 receptor extracellular domain fused to IgG contant regions
  • the receptor in an immobilised form, e.g. via an affinity column.
  • Binding may be assessed quantitatively or qualitatively.
  • the method may comprise determining the difference in binding of the TREM-1 ligand or derivative to the TREM-1 receptor or derivative in the absence of the test compound with that occurring in the presence of the test compound.
  • a qualitative assay may simply determine whether or not binding has occurred.
  • the binding may be detected through use of a competitive immunoassay, a non-competitive assay system using techniques such as western blots, a radioimmunoassay, an ELISA (enzyme linked immunosorbent assay), a "sandwich” immunoassay, an immunoprecipitation assay, a precipitin reaction, a gel diffusion precipitin reaction, an immunodiffusion assay, an agglutination assay, a complement fixation assay, an immunoradiometric assay, a fluorescent immunoassay, a protein A immunoassay, an immunoprecipitation assay, an immunohistochemical assay, a competition or sandwich ELISA, a radioimmunoassay, a Western blot assay, an immunohistological assay, an immunocytochemical assay, a dot blot assay, a fluorescence polarisation assay, a scintillation proximity
  • Suitable techniques use a detectable label and measure changes in the amount of label detected.
  • CD177 (sometimes known as NB1 or PRV-1 ) as a TREM-1 ligand. They have also shown that a monoclonal antibody to CD177 blocks the binding of constructs comprising the TREM-1 ligand to septic neutrophils expressing the TREM-1 receptor.
  • CD177 was well known prior to the present invention, but there was nothing to indicate that it was a TREM-1 ligand. Indeed there was no discussion at all of CD177 in connection with TREM-1 ligands.
  • CD177 is discussed in connection with autoimmune disorders. For example it is explained in Stroneck et al in Transl Med. 2004; 2: 8, that CD177is a neutrophil membrane glycoprotein that was first described by Lalezari et al while investigating a case of neonatal alloimmune neutropenia [Lalezari P, Murphy GB, Allen FH Jr. NB1 , a new neutrophil-specific antigen involved in the pathogenesis of neonatal neutropenia J CHn Invest. 1971 ; 50:1108-1 115)]. Occasionally, during pregnancy, a mother produces alloantibodies to neutrophil antigens than cross the placenta, react with neutrophils in the fetus, and cause the neonate to become neutropenic.
  • NB1 Human Neutrophil Antigen-2a
  • HNA-2a Human Neutrophil Antigen-2a
  • gp carrying this antigen was called NB1 gp [Bux J, Bierling P, von dem Borne AEG Kr, et al. ISBT Granulocyte Antigen Working Party. Nomenclature of Granulocyte Alloantigens. Vox Sang. 1999; 77:251].
  • Temerinac and colleagues identified and sequenced PRV-1 in 2000 while searching for genes overexpressed in neutrophils from patients with polycythemia vera [Temerinac S, Klippel S, Strunck E, Roder S, Lubbert M, Lange W, Azemar M, Meinhardt G, Schaefer HE, Pahl HL. Cloning of PRV-1 , a novel member of the uPAR receptor superfamily, which is overexpressed in polycythemia rubra vera. Blood. 2000; 95:2569-2576].
  • NB1 and PRV-1 differ at only 4 nucleotides that result in amino acid changes and Caruccio, Bettinotti, and colleagues have shown that PRV-1 and NB1 are alleles of a single gene [Bettinotti MP, Olsen A, Stroncek D. The Use of Bioinformatics to Identify the Genomic Structure of the Gene that Encodes Neutrophil Antigen NB1 , CD177. Clinical Immunology. 2002; 102:138-144; Caruccio L, Walkovich K, Bettinotti M, Schuller R, Stroncek D. CD177 polymorphisms: correlation between high frequency single nucleotide polymorphisms and neutrophil surface protein expression. Transfusion. 2004; 44:77-82].
  • PRV-1 and NB1 are now considered to be alleles of the same gene, with PRV-1 being the more common allele in a normal population [Caruccio L, Bettinotti M, Fraser E, Director-Myska A, Arthur DC, Stroncek DF Blood. 2003;102:661 a].
  • derivatives of CD177 can be used in the present invention.
  • derivative includes variants, fragments and fusion proteins.
  • Suitable derivatives bind to a TREM-1 receptor under physiological conditions. More suitably, they do not bind to any other cell surface protein present in vivo upon neutrophils or monocytes, especially to neutrophils or monocytes from septic patients. This enables them to be used in cell-based binding assays that are highly specific.
  • derivatives are specific for a TREM-1 receptor in the sense that they do not bind to any other protein that is normally found in the species (e.g. Homo sapiens) from which the receptor is obtained.
  • Variants of CD177 include allelic variants.
  • Allelic variant may be intra-species or inter-species allelic variants. Suitable variants occur in mammals. More suitably they occur in rodents (e.g. mice, rats) or rabbits or in humans.
  • Non-allelic variants are also included.
  • Such molecules can be prepared using recombinant DNA technology, automated synthesis, site directed mutagenesis, etc. Such techniques are now well developed.
  • Suitable variants have an amino acid sequence (or at least a part thereof) that is at least about 60%, 70%, 75%, 80%, 85%, 90%, 95%, or 98% identical to the amino acid sequence shown in Figure 18, or at least to a part of the Figure 18 sequence that is required for binding to the TREM- 1 receptor for CD177.
  • This part is expected to be within a fragment corresponding to amino acids from 22 to 437 especially from 22 to 408 shown in Figure 18.
  • This stretch of amino acids or (even a smaller part thereof that still binds to the CD177 receptor) can be used in the present invention and can also be used for sequence comparisons.
  • the amino acid sequence 1 to 22 shown in Figure 18 will not normally be present in the mature protein.
  • Amino acid 408 is the amino acid used for attachment to the GPI anchor.
  • An enzyme that cleaves the GPI anchor e.g. Phospolipase C
  • Other soluble forms are also possible and can be made by genetic engineering, as discussed in greater detail later on.
  • sequences are aligned for optimal comparison purposes (e.g., gaps may optionally be introduced in one or both of a first and a second amino acid or nucleic acid sequence for optimal alignment and non-homologous sequences can be disregarded for comparison purposes).
  • the length of a reference sequence aligned for comparison purposes is at least 30%, suitably at least 40%, more suitably at least 50%, even more suitably at least 60%, and even more suitably at least 70%, 80%, or 90% of the length of the reference sequence, such as the whole of the length of the reference sequence (e.g. when aligning a second sequence to the first amino acid sequence which has for example 100 amino acid residues, at least 30, suitably at least 40, more suitably at least 50, even more suitably at least 60, and even more suitably at least 70, 80 or 90 amino acid residues are aligned, such as 100 amino acid residues).
  • the amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared.
  • amino acid or nucleic acid “identity” is equivalent to amino acid or nucleic acid “homology”
  • the percentage identity between the two sequences is a function of the number of identical positions shared by the sequences, taking into account the number of gaps, and the length of each gap, which need to be introduced for optimal alignment of the two sequences.
  • the comparison of sequences and determination of percent identity between two sequences can be accomplished using a mathematical algorithm.
  • the percentage identity between two amino acid sequences is determined using the Needleman and Wunsch (J. MoI. Biol. (48):444-453 (1970)) algorithm which has been incorporated into the GAP program in the GCG software package (available at http://www.gcg.com), using either a Blossom 62 matrix or a PAM250 matrix, and a gap weight of 16, 14, 12, 10, 8, 6, or 4, and a length weight of 1 , 2, 3, 4, 5, or 6.
  • the percent identity between two nucleotide sequences is determined using the GAP program in the GCG software package (available at http://www.gcg.com), using a NWSgapdna.CMP matrix and a gap weight of 40, 50, 60, 70, or 80, and a length weight of 1 , 2, 3, 4, 5, or 6.
  • the percentage identity between two amino acid or nucleotide sequences is determined using the algorithm of E. Meyers and W. Miller (CABIOS, 4:1 1-17 (1989)) which has been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4.
  • the nucleic acid and protein sequences of the present invention can further be used as a "query sequence" to perform a search against public databases to identify, for example, other family members or related sequences.
  • search can be performed using the NBLAST and XBLAST programs (version 2.0) of Altschul, et al. (1990) J. MoI. Biol. 215:403-10.
  • Gapped BLAST can be utilized as described in Altschul et al., (1997) Nucleic Acids Res. 25(17):3389-3402.
  • the default parameters of the respective programs e.g., XBLAST and NBLAST
  • XBLAST and NBLAST See http://www.ncbi.nlm.nih.gov.
  • amino acids glycine, alanine, valine, leucine and isoleucine can often be substituted for one another (amino acids having aliphatic side chains).
  • amino acids having aliphatic side chains amino acids having aliphatic side chains.
  • glycine and alanine are used to substitute for one another (they have relatively short side chains) and that valine, leucine and isoleucine are used to substitute for one another (they have larger aliphatic side chains which are hydrophobic).
  • amino acids that can often be substituted for one another typically include: phenylalanine, tyrosine and tryptophan (amino acids having aromatic side chains); lysine, arginine and histidine (amino acids having basic side chains); aspartate and glutamate (amino acids having acidic side chains); asparagine and glutamine (amino acids having amide side chains); and cysteine and methionine (amino acids having sulphur containing side chains). Substitutions of this nature are often referred to as “conservative” or “semi-conservative" amino acid substitutions.
  • the variant may contain 10 or fewer substitutions (e.g. 5 or fewer, more suitably 1 or 2).
  • Deletions of inessential or undesired parts of a polypeptide can be made. This can be useful in reducing the size of a polypeptide. As discussed later, deletions can also be useful in producing soluble polypeptides if a polypeptide is normally membrane bound.
  • the variant may contain one or two deletions, each of which is 20% or less (such as 10% or less) of the length of the reference sequence.
  • Amino acid insertions can also be made. This may be done to alter the properties of the polypeptide (e.g. to assist in identification, purification or expression).
  • the variant may contain one or two insertions, each of which is each of which is 20% or less (such as 10% or less) of the length of the reference sequence.
  • Polypeptides incorporating amino acid changes can be provided using any suitable techniques.
  • a nucleic acid sequence incorporating a sequence change can be provided by site directed mutagenesis. This can then be used to allow the expression of a polypeptide having a corresponding change in its amino acid sequence.
  • polypeptides comprising one or more amino acid analogues (including non-naturally occurring amino acids) may be used.
  • the present inventions includes mimetopes and peptidomimetics.
  • mimetope and peptidomimetic are used interchangeably herein.
  • a “mimetope” of a compound X refers to a compound in which chemical structures of X necessary for functional activity of X have been replaced with other chemical structures which mimic the conformation of X.
  • peptidomimetics include peptidic compounds in which the peptide backbone is substituted with one or more benzodiazepine molecules (see e.g., James, G. L. et al.
  • amino acid mimetics include D-amino acids. Peptides substituted with one or more D-amino acids may be made using well known peptide synthesis procedures.
  • Additional substitutions include amino acid analogues having variant side chains with functional groups, for example, b-cyanoalanine, canavanine, djenkolic acid, norleucine, 3-phosphoserine, homoserine, dihydroxyphenylalanine, 5-hydroxytryptophan, 1-methylhistidine, or 3-methylhistidine.
  • Methods for preparing mimetopes and peptidomimetics are known in the art.
  • fragments may be utilised in the screening methods. They may also be used for other purposes e.g. for raising antibodies, for binding studies; for therapeutic purposes (as discussed later), etc.
  • Fragments suitably include at least 10, 20, 30, 40, 50, 60, 70, 80, 90 or 100 amino acids of the amino acid sequence shown in Figure 18 or of variants thereof.
  • Suitable fragments are soluble forms.
  • the term "soluble" is used herein to distinguish from a polypeptide that is membrane-bound.
  • a soluble form can be made by cleaving a GPI anchored protein with a suitable enzyme, as discussed earlier.
  • genetic engineering techniques may be used to provide a protein that is secreted and does not have a GPI anchor.
  • Different lengths of soluble forms can be provided and can be derived from the extracellular portion of a TREM-1 ligand or variant.
  • TREM-1 receptor binding portion is present, as can be easily determined by binding studies.
  • variants or fragments described above may be linked with heterologous moieties (i.e. moieties with which they are not normally linked in nature).
  • link is via a covalent bond, although non-covalent linkages are also within the scope of the present invention.
  • fusion proteins may be provided.
  • the present invention encompasses fusion proteins in which the TREM-1 ligands or derivatives thereof (especially fragments) are recombinantly fused or chemically conjugated (including both covalent and non-covalent conjugations) to heterologous polypeptides (i.e., an unrelated polypeptide or portion thereof, suitably at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90 or at least 100 amino acids of the polypeptide) to generate fusion proteins.
  • heterologous polypeptides i.e., an unrelated polypeptide or portion thereof, suitably at least 10, at least 20, at least 30, at least 40, at least 50, at least 60, at least 70, at least 80, at least 90 or at least 100 amino acids of the polypeptide
  • the fusion does not necessarily need to be direct, but may occur through linker sequences.
  • fusion is to sequences derived from various types of immunoglobulins.
  • fusion can be to a constant region (e.g., hinge, CH2, and CH3 domains) of human IgGI or IgM molecule, (for example, as described by Hudson & Souriauso (2003) Nature Medicine 9(1 ):129 - 134) so as to make the fused polypeptides or fragments thereof more soluble and stable in vivo.
  • the short half-life of antibody fragments can also be extended by 'pegylation', that is, a fusion to polyethylene glycol (see Leong, S. R. et al. (2001 ) Cytokine 16:106-119).
  • Fc domains are fused with biologically active peptides.
  • a pharmacologically active compound is produced by covalently linking an Fc domain to at least one amino acid of a selected peptide. Linkage to the vehicle increases the half- life of the peptide, which otherwise could be quickly degraded in vivo
  • fusion proteins comprising non-classical alternative protein scaffolds can be made (for example see Nygren & Skerra (2004) J Immunol Methods 290(1 -2):3-28 or WO03049684).
  • Such fusion proteins can be used as an immunogen for the production of specific antibodies which recognize the polypeptides of the invention or fragments thereof.
  • fusion proteins can be administered to a subject so as to inhibit interactions between the TREM-1 ligand and its receptor in vivo.
  • N-terminal signal sequence fusions to signal sequences can be provided if desired.
  • Various signal sequences are commercially available.
  • the secretory sequences of melittin and human placental alkaline phosphatase (Stratagene; La JoIIa, CA) are available as eukaryotic heterologous signal sequences.
  • the phoA secretory signal (Sambrook, et al., supra; and Current Protocols in Molecular Biology, 1992, Ausubel, et al., eds., John Wiley & Sons) and the protein A secretory signal (Pharmacia Biotech; Piscataway, NJ) can be listed.
  • Another example is the gp67 secretory sequence of the baculovirus envelope protein (Current Protocols in Molecular Biology, 1992, Ausubel, et al., eds., John Wiley & Sons).
  • fusion is to tag sequences, e.g., a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 9131 1 ), among others, many of which are commercially available.
  • a hexa-histidine peptide such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 9131 1 ), among others, many of which are commercially available.
  • pQE vector QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 9131 1
  • hexa-histidine provides for convenient purification of the fusion protein.
  • peptide tags are the hemagglutinin "HA” tag, which corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson, et al., 1984, Cell 37:767) and the "flag” tag (Knappik, et al., 1994, Biotechniques 17(4)754-761 ). These tags are especially useful for purification of recombinantly produced polypeptides.
  • Fusion proteins can be produced by standard recombinant DNA techniques or by protein synthetic techniques, e.g., by use of a peptide synthesizer.
  • a nucleic acid molecule encoding a fusion protein can be synthesized by conventional techniques including automated DNA synthesizers.
  • PCR amplification of gene fragments can be carried out using anchor primers which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed and reamplified to generate a chimeric gene sequence (see, e.g., Current Protocols in Molecular Biology, 1992, Ausubel, et al., eds., John Wiley & Sons).
  • the nucleotide sequence coding for a fusion protein can be inserted into an appropriate expression vector.
  • Suitable fusions proteins are multivalent in the sense that they have a plurality of parts (or ligands) that can bind to a TREM-1 receptor.
  • soluble CD177 ligands i.e. proteins capable of specifically binding to CD177
  • Molecules such as immunoglobulins can be used to provide convenient multimeric scaffolds.
  • a dimer based upon fusing a TREM-1 ligand coding region with a region encoding a mutated form of the Fc portion of IgG is discussed in the examples (the Fc portion is modified so that it does not bind to Fc receptors).
  • the dimer is produced upon association of the Fc regions once the fusion proteins polypeptides have been secreted into a cell culture medium.
  • the scaffold may be provided by any desired structure.
  • streptavidin can be used. As discussed earlier this has been successfully by the present inventors as a scaffold to provide a tetramer of the TREM-1 receptor. A similar technique can be used to provide a tetramer for the TREM-1 ligand or derivative thereof. Different multimers (e.g. dimers, trimers, tetramers, heptamers, hexamers, etc.) may be provided by linking different numbers of TREM- 1 ligands/derivatives thereof to an appropriate scaffold.
  • multimers e.g. dimers, trimers, tetramers, heptamers, hexamers, etc.
  • the scaffold it is desired that it does not substantially interfere with the binding to the TREM-1 receptor.
  • Suitable structures are at least as capable of binding to the TREM-1 receptor as is the wild-type TREM-1 ligand. More suitably they have a higher probability of binding to the TREM-1 receptor.
  • the scaffold does not significantly increase inflammation in a mammalian (suitably human) host. Proteins that are already present in a given species can be used as the basis for suitable scaffolds, given that these are less likely to provoke immune responses.
  • a TREM-1 ligand, fragment or variant need not however be linked to another polypeptide, as in the case of a fusion protein, but can be linked to a surface.
  • Immobilised forms may be used for many purposes, including purification, diagnosis, screening (especially high throughput screening), characterisation, storage, ease of handling, etc.
  • TREM-1 ligand or derivatives thereof can be used in the present invention, including variants, fragments, fusion proteins, etc.
  • the ligand or derivative may be provided in "isolated" form.
  • an "isolated" polypeptide is considered to be substantially free of cellular material or other contaminating polypeptides from the cell or tissue source from which the protein is derived, or is substantially free of chemical precursors or other chemicals when chemically synthesised.
  • substantially free of cellular material includes preparations of a polypeptide in which the polypeptide is separated from cellular components of the cells from which it is isolated or recombinantly produced.
  • a polypeptide/protein that is substantially free of cellular material includes preparations of the polypeptide/protein having less than about 50%, 40%, 30%, 20%, 10%, 5%, 2.5%, or 1 %, (by dry weight) of contaminating protein.
  • the polypeptide When the polypeptide is recombinantly produced, it is also suitably substantially free of culture medium, i.e. the culture medium represents less than about 50%, 40%, 30%, 20%, 10%, or 5% of the volume of the protein preparation.
  • the polypeptide When the polypeptide is produced by chemical synthesis, it is suitably substantially free of chemical precursors or other chemicals, i.e., it is separated from chemical precursors or other chemicals which are involved in the synthesis of the protein. Accordingly, such preparations of the polypeptide/protein have less than about 50%, 40%, 30%, 20%, 10%, or 5% (by dry weight) of chemical precursors or compounds other than polypeptide fragment of interest.
  • Isolated forms may be used for analysis of structure/function, for binding studies, for screening, for raising or selecting antibodies, etc. The fact that they there is relatively little or no contamination with other proteins means that results are unlikely to be adversely affected by contamination.
  • the invention includes the use of a compound that blocks or reduces the binding of a TREM-1 ligand to the TREM-1 receptor in the manufacture of a medicament for treating a disorder that is characterised by the release of one or more proinflammatory cytokines or chemokines.
  • the disorder may be any inflammatory disorder (or other disorder) that is mediated by the binding of the TREM-1 ligand to a TREM-1 receptor.
  • inflammatory disorders include (but are not limited to) acute and chronic inflammatory disorders, sepsis, acute endotoxemia, encephalitis, Inflammatory Bowel Disease (IBD), Chronic Obstructive Pulmonary Disease (COPD), allergic inflammatory disorders, asthma, pulmonary fibrosis, pneumonia, Community acquired pneumonia (CAP), Ventilator associated pneumonia (VAP), Acute respiratory infection, Acute respiratory distress syndrome (ARDS), Infectious lung diseases, Pleural effusion, Peptic ulcer, Helicobacter pylori infection, hepatic granulomatosis, arthritis, rheumatoid arthritis, osteoarthritis, inflammatory osteolysis, ulcerative colitis, psoriasis, vasculitis, autoimmune disorders, thyroiditis, Meliodosis, (mesenteric) Ischemia reperfusion, Filovirus infection, Infection of the urinar
  • TREM-1 signalling is implicated in diseases in which monocyte-platelet and neutrophil-platelet aggregates play an important role (Haselmayer et al. Blood 2007 110:1029- 1035).
  • circulating leukocyte-platelet aggregates especially monocyte-platelet aggregates, promote the formation of atherosclerotic lesions, are increased in acute coronary syndromes, stroke, and peripheral vascular disease, and are an early marker of acute myocardial infarction.
  • the disorder is sepsis and is mediated by a pathogen.
  • An example of sepsis that is microbially mediated is pneumonia.
  • the disorder is Inflammatory Bowel Disease.
  • sTREM-1 means, an inflammation of the lung caused by infection by extracellular pathogens such as bacterial infection, and non-bacterial infections (for example, infection by Blastomyces dermatitidis, Histoplasma capsulatum, Coccidioides, Sporothrix schenckii, Pneumocystis carinii, Cryptococcus, Aspergillus, or Mucor sp.), protozoal infections or parasitic infections (for example, those caused by Toxoplasma gondii, Strongyloides stercoralis, Ascaris, hookworm, Dirofilaria, Paragonimus, or Entamoeba histolytica) where increased expression of sTREM-1 can be detected.
  • non-bacterial infections for example, infection by Blastomyces dermatitidis, Histoplasma capsulatum, Coccidioides, Sporothrix schenckii, Pneumocystis carinii, Cryptococcus, Aspergill
  • Pneumonia includes "Lobar Pneumonia” (which occurs in one lobe of the lung) and Bronchopneumonia (tends to be irregularly located in the lung). Furthermore, pneumonia is often classified into two categories that may help predict the organisms that are the most likely culprits. "Community-acquired (pneumonia contracted outside the hospital). Pneumonia" in this setting often follows a viral respiratory infection. It affects nearly 4 million adults each year. It is likely to be caused by Streptococcus pneumoniae, the most common pneumonia-causing bacteria. Other organisms, such as atypical bacteria called Chlamydia or Mycoplasma pneumonia are also common causes of community-acquired pneumonia. "Hospital-acquired pneumonia" contracted within the hospital is often called nosocomial pneumonia. Hospital patients are particularly vulnerable to gram-negative bacteria and staphylococci.
  • a wide range of compounds can be used in treatment of the above-mentioned disorders.
  • Such a compound is an antibody.
  • the antibody binds to the TREM-1 ligand (or variant, fragment or fusion proteins thereof as appropriate).
  • TREM-1 ligand that is responsible for binding to the TREM-1 receptor.
  • the antibody may be monoclonal or polyclonal.
  • Polyclonal antibodies can be raised by stimulating their production in a suitable animal host (e.g. a mouse, rat, guinea pig, rabbit, sheep, goat or monkey) when a TREM-1 ligand or derivative as immunogen is injected into the animal. If necessary, an adjuvant may be administered together with the substance of the present invention. The antibodies can then be purified by virtue of their binding to a substance of the present invention.
  • a suitable animal host e.g. a mouse, rat, guinea pig, rabbit, sheep, goat or monkey
  • an adjuvant may be administered together with the substance of the present invention.
  • the antibodies can then be purified by virtue of their binding to a substance of the present invention.
  • the immunogen may be CD177 or a fragment or variant thereof.
  • the immunogen may be cells expressing a TREM-1 ligand, such as neutrophils expressing a TREM-1 ligand such as CD177.
  • the immunogen is of human type (eg human CD177 or human cells expressing TREM-1 ligand).
  • Monoclonal antibodies can be produced from hybridomas. These can be formed by fusing myeloma cells and spleen cells from animals which produce the desired antibody in order to form an immortal cell line. Thus the well-known Kohler & Milstein technique (Nature, 256, 52-55 (1975)) or variations upon this technique can be used.
  • antibody is used herein to include derivatives thereof which are capable of binding to polypeptides of the present invention.
  • the present invention includes antibody fragments and synthetic constructs. Examples of antibody fragments and synthetic constructs are given by Dougall et al in Tibtech 12 372-379 (September 1994).
  • Antibody fragments include, for example, Fab, F(ab') 2 and Fv fragments.
  • Fv fragments can be modified to produce a synthetic construct known as a single chain Fv (scFv) molecule. This includes a peptide linker covalently joining V h and Vi regions, which contribute to the stability of the molecule.
  • Other synthetic constructs which can be used include CDR peptides. These are synthetic peptides comprising antigen-binding determinants. Peptide mimetics may also be used. These molecules are usually conformationally restricted organic rings which mimic the structure of a CDR loop and which include antigen-interactive side chains.
  • Synthetic constructs include chimeric antibodies.
  • one or more parts of an antibody are derived from one animal (usually rodent) and one or more parts from another animal (usually humans).
  • rodent one or more parts from another animal (usually humans).
  • Preferred expression systems are mammalian cell cultures (e.g. CHO cells)
  • Preferred chimeric antibodies are humanised antibodies, sometimes known as CDR grafted antibodies. These are alternatives to more traditional chimeric antibodies. Here only the complimentarity determining regions from non-human (usually rodent) antibody V-regions are combined with framework regions from human V-regions. These antibodies are considered to be less immunogenic than older style chimeric antibodies, where the whole of the variable regions are derived from non-human animals. Thus undesired side effects are less likely.
  • Completely human antibodies can also be produced. For ethical reasons it is not desirable to produce these directly from humans. However they can be made by a variety of methods known in the art including phage display using antibody libraries derived from human immunoglobulin sequences. (See U.S. Patent Nos. 4,444,887 and 4,716,11 1 ; and PCT publications WO 98/46645; WO 98/50433; WO 98/24893; WO 98/16654; WO 96/34096; WO 96/33735; and WO 91/10741.). Human antibodies can also be produced using transgenic mice (see Lonberg and Huszar (1995), Int. Rev. Immunol. 13:65-93).
  • any of the aforesaid antibodies/constructs with an additional moiety which provides the molecule with some desirable property in addition to antigen binding.
  • the moiety may be a detectable label, a compound that increases the stability/half life of the antibody, in vivo etc.
  • Modifications such as lipidation can be used to stabilize antibodies and to enhance uptake and tissue penetration (e.g., into the brain).
  • a method for lipidation of antibodies is described by Cruikshank, et al., 1997, J. Acquired Immune Deficiency Syndromes and Human Retrovirology 14:193). Reference can also be made to Leong, S. R. et al. (2001 ) Cytokine 16:106-1.
  • the half-life of antibody fragments can also be extended by 'pegylation', that is, by fusion to polyethylene glycol.
  • a monoclonal antibody is used (e.g. the antibody R33).
  • aspects of the invention include: a method for obtaining anti-TREM-1 ligand antibodies comprising providing a TREM-1 ligand or a derivative thereof and using it to generate antibodies in a non-human host, e.g. by immunising said non-human host (eg rabbit or rodent, such as mouse or rat) with TREM-1 ligand or a derivative thereof as immunogen; also a method for obtaining anti-TREM-1 ligand antibodies comprising providing cells such as neutrophils, which present on their surface a TREM-1 ligand or a derivative thereof and using them to generate antibodies in a non-human host e.g. by immunising said non-human host (eg rabbit or rodent, such as mouse or rat) with cells such as neutrophils, which present on their surface a TREM-1 ligand or a derivative thereof as immunogen.
  • a method for obtaining anti-TREM-1 ligand antibodies comprising providing a TREM-1 ligand or a derivative thereof and using it to generate antibodies in
  • An alternative way of blocking or reducing binding of the TREM-1 ligand to a TREM-1 receptor is to use a soluble form of the TREM-1 ligand or a soluble variant thereof, e.g. a multimer as described earlier.
  • TREM-1 ligand it is also possible to block or reduce the expression of the TREM-1 ligand, rather than rely upon blocking the binding of the ligand to the receptor. This can be done by blocking or reducing transcription or by blocking or reducing translation.
  • transcriptional blockers of the gene for the TREM-1 ligand or down-regulators may be provided.
  • the gene for the TREM-1 ligand may be inactivated (e.g. by using targeted homologous recombination techniques to disrupt the gene / promoter).
  • antisense molecules may be provided. These may hybridise to TREM-1 RNA so as to prevent or reduce translation thereof. Suitably hybridisation is specific so that there is not significant hybridisation to different RNA molecules produced naturally by neutophils or monocytes in vivo.
  • Hybridisation can be tested in vitro if desired.
  • stringent conditions can be provided and it can then be determined whether or not hybridisation occurs.
  • Suitable antisense molecules hybridise under stringent conditions.
  • stringent hybridisation conditions involves using a pre-washing solution of 5 X SSC, 0.5% SDS, 1.OmM EDTA (pH 8.0) and attempting hybridisation overnight at 55°C using 5 X SSC.
  • 5 X SSC 0.5% SDS
  • 1.OmM EDTA pH 8.0
  • Hybridisation conditions are discussed in detail at pp 1.101 -1.1 10 and 11.45 - 11.61 of Sambrook et al [Molecular Cloning, 2nd Edition, Cold Spring Harbor Laboratory Press (1989)].
  • Antisense molecules can be introduced via a suitable vehicle (e.g. a liposome). They may even be introduced directly e.g. by using gene gun technology. Alternatively a vector may be provided that produces such molecules in vivo.
  • a suitable vehicle e.g. a liposome
  • a vector may be provided that produces such molecules in vivo.
  • RNA interference double stranded RNA molecules that partake in RNA interference (RNAi) may also be used.
  • targeted RNA is physically cleaved and therefore the mechanism of action of RNAi is quite different from that of antsisense molecules that act simply by binding to RNA so that it is no longer available for translation.
  • Andrew Fire and Craig C. MeIIo shared the Nobel Prize in Physiology or Medicine for their work on RNA interference in the nematode worm C. elegans, [Fire A, Xu S, Montgomery M, Kostas S, Driver S, MeIIo C (1998). "Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans".
  • RNAi RNAi-binding protein
  • Relevant papers include: Dorsett, Y and Tuschl, T (2004). siRNAs: Applications in functional genomics and potential as therapeutics. Nature Reviews 3, 318-329; Hannon, GJ and Rose, JJ (2004). Unlocking the potential of the human genome with RNA interference. Nature 431 , 371-378; Soutschek, J et al. (2004). Therapeutic silencing of an endogenous gene by systematic administration of modified siRNAs. Nature 432, 173-178; Morrisey, DV et al. (2005). Potent and persistent in vivo anti-HBV activity of chemically modified siRNAs. Nat.
  • Ribozymes may also be used. These are single stranded RNA molecules (usually with double stranded hairpin regions) that have enzymatic activity. Ribozymes can be engineered that bind to and cleave target RNA molecules. This is discussed for example by Citti and Rainaldi in Curr Gene Ther. 2005 Feb;5(1 ):11-24 "Synthetic hammerhead ribozymes as therapeutic tools to control disease genes".
  • the compound may be administered as a pharmaceutical composition together with a pharmaceutically acceptable acceptable diluent, carrier or excipient.
  • pharmaceutically acceptable diluent, carrier or excipient is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions.
  • the invention includes methods for preparing pharmaceutical compositions containing a peptide or polypeptide of the invention. Such compositions can further include additional active agents. Thus, the invention further includes methods for preparing a pharmaceutical composition by formulating a pharmaceutically acceptable carrier with a peptide or polypeptide of the invention and one or more additional active compounds.
  • a pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, transdermal (topical), transmucosal, intra-articular, intraperitoneal, and intrapleural, as well as oral, inhalation, and rectal administration.
  • Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose. pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • suitable carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF; Parsippany, NJ) or phosphate buffered saline (PBS).
  • the composition must be sterile and should be fluid to the extent that easy injectability with a syringe exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, sodium chloride in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by incorporating the active compound (e.g., a polypeptide or antibody) in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle which contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the more suitable methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Oral compositions generally include an inert diluent or an edible carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches, or capsules. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition.
  • the tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient, such as starch or lactose; a disintegrating agent, such as alginic acid, Primogel, or corn starch; a lubricant, such as magnesium stearate or Sterotes; a glidant, such as colloidal silicon dioxide; a sweetening agent, such as sucrose or saccharin; or a flavouring agent, such as peppermint, methyl salicylate, or orange flavouring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose
  • a disintegrating agent such as alginic acid, Primogel, or corn starch
  • a lubricant such as magnesium stearate or Sterotes
  • a glidant such as colloidal silicon dioxide
  • the compounds are delivered in the form of an aerosol spray from a pressurized container or dispenser which contains a suitable propellant, e.g. a gas such as carbon dioxide, or a nebuliser.
  • a suitable propellant e.g. a gas such as carbon dioxide, or a nebuliser.
  • Systemic administration can also be by transmucosal or transdermal means.
  • penetrants appropriate to the barrier to be permeated are used in the formulation.
  • penetrants are generally known in the art, and include, for example, for transmucosal administration, detergents, bile salts, and fusidic acid derivatives.
  • Transmucosal administration can be accomplished through the use of nasal sprays or suppositories.
  • the active compounds are formulated into ointments, salves, gels, or creams as generally known in the art.
  • the compounds can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery.
  • the active compounds are prepared with carriers that will protect the compound against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • a controlled release formulation including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc.
  • Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Patent No. 4,522,811.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved, and the limitations inherent in the art of compounding such an active compound for the treatment of individuals.
  • a therapeutically effective amount of a polypeptide suitably ranges from about 0.001 to 30 mg/kg body weight, suitably about 0.01 to 25 mg/kg body weight, more suitably about 0.1 to 20 mg/kg body weight, and even more suitably about 1 to 10 mg/kg, 2 to 9 mg/kg, 3 to 8 mg/kg, 4 to 7 mg/kg, or 5 to 6 mg/kg body weight.
  • a suitable dosage is 0.1 mg/kg to 100 mg/kg of body weight (generally 10 mg/kg to 20 mg/kg). If the antibody is to act in the brain, a dosage of 50 mg/kg to 100 mg/kg is usually appropriate. Generally, partially human antibodies and fully human antibodies have a longer half-life within the human body than other antibodies. Accordingly, lower dosages and less frequent administration is often possible.
  • the actual dosage will be determined by a physician. If desired, a low starting dosage can be used and can gradually be increased until a beneficial effect is obtained. If side effects develop, then the dosage can be reduced in accordance with normal clinical practice.
  • compositions can be included in a container, pack, or dispenser together with instructions for administration.
  • the present invention also has various diagnostic applications.
  • the disorder may be any of the disorders discussed earlier in respect of medical uses.
  • the invention includes a method comprising obtaining a biological sample and analysing the sample for a TREM-1 ligand or for TREM-1 ligand mRNA.
  • the sample can be a sample of whole blood, blood serum, blood plasma, urine, a cellular fraction of blood, a tissue sample, etc.
  • the sample is suitably a sample comprising cells (suitably neutrophils and/or monocytes) if it is desired to analyse membrane bound TREM-1 ligand. Similarly cells will normally be used if it is desired to analyse mRNA
  • the sample is suitably a sample comprising extracellular fluid (e.g. serum, plasma or urine) if it is desired to analyse soluble TREM-1 ligand. This is because the soluble form is shed into extracellular fluid.
  • extracellular fluid e.g. serum, plasma or urine
  • the sample will normally be taken from a patient thought to have, or to be at risk of having, any of the disorders discussed earlier.
  • the presence or absence of the ligand or of corresponding mRNA may simply be identified. This can be useful if this is not present at all in a healthy individual or is present only at very low levels that are difficult to detect.
  • the method includes a step of quantifying the TREM-1 ligand or TREM-1 ligand mRNA in the biological sample.
  • It may further comprise a negative control comparing the level of the ligand or corresponding RNA with a control level or range corresponding to what would be expected for a healthy individual.
  • TREM-1 ligand or TREM-1 ligand mRNA may be significantly above that of the control this may be an indicator that an individual is likely to have the disorder
  • It may comprise a positive control, comprising comparing the level of the ligand or corresponding mRNA with a control level or range corresponding to what would be expected from an individual having the disorder.
  • TREM-1 ligand or TREM-1 ligand mRNA may then be an indication that an individual is likely to have the disorder
  • the method may for example use an antibody to the ligand.
  • a suitable antibody is specific to the ligand. It may be a monoclonal antibody (e.g. R33).
  • a nucleic acid molecule that hybridises to the TREM-1 mRNA can be used (e.g. a probe or primer).
  • the mRNA may be used to generate cDNA and a nucleic acid molecule that hybridises to the cDNA may be used. If desired amplification techniques such as reverse PCR can be used, although these are not essential.
  • nucleic acid molecule is capable of hybridising under stringent conditions, as described earlier.
  • a further method that is useful in diagnosis is to provide a soluble form of the TREM-1 ligand or soluble variant thereof to bind to the TREM-1 receptor. This can be used to detect the TREM-1 receptor and/or to quantify the amount of receptor present in a sample.
  • the present invention also provides diagnostic kits.
  • kits for diagnosing a disorder that is characterised by the release in vivo of one or more pro-inflammatory cytokines or chemokines wherein the kit comprises a compound that binds to a TREM-1 ligand or to DNA or RNA encoding said ligand.
  • the kit may comprises a TREM-1 ligand or a variant thereof (suitably a soluble form) that binds to the TREM-1 receptor.
  • the compound may for example be an antibody that binds to the TREM-1 ligand or a nucleic acid that hybridises to TREM-1 RNA or DNA.
  • the kit suitably includes comprising means for detecting and/or quantifying the binding.
  • This means may for example be one or more indicators that provide a visible change if said disorder is present.
  • the indicator(s) may for example provide a colour change or a change in marking.
  • the kit may itself include one or more controls.
  • controls comprising biological samples from healthy patients.
  • the samples may comprise cells (e.g. neutrophils and/or monocytes), as discussed earlier.
  • TREM-1 ligand may be cell free.
  • serum, plasma or urine samples may be provided if it is desired to screen to look for soluble forms of the TREM-1 ligand.
  • the controls need not even be physical samples. They may simply be indicators of what would be expected for healthy patients. Such indicators can be provided on instructions, packaging, labelling, etc. They may be in the form of charts, figures, ranges, etc.
  • Components of the kit may be enclosed within different containers, which may be sealed and may be in sterile form.
  • the containers may be within a package for the kit, along with instructions for determining whether a subject is at risk of developing a disorder as described previously.
  • kits for identifying the presence of a mutant form of a TREM-1 ligand in addition to the aforesaid kits, also includes kits for identifying the presence of a mutant form of a TREM-1 ligand.
  • mutant form is used herein to distinguish from the most common form known in nature in a given species, which is usually known as the "wild type". Thus in the case of a gene encoding a mutant form, this may will differ from a gene encoding the wild type form by one or more coding nucleotides. In the case of a polypeptide a mutant form may differ by one or more amino acids. Mutant forms therefore include allelic variants.
  • kit may, for example, comprise an antibody that binds more strongly to a mutant form than to a wild type form of the ligand, or may comprise a nucleic acid that binds more strongly to a nucleic acid encoding the mutant form than to a nucleic acid encoding the wild type form.
  • the kit may comprise a control allowing binding to be compared with binding to the wild type ligand or to a nucleic acid encoding the wild type ligand.
  • the antibody may be specific for the mutant form of the TREM-1 ligand.
  • the nucleic acid may be specific (under stringent hybridisation conditions) for a nucleic acid encoding the mutant form of the TREM-1 ligand.
  • Mutant forms are of interest because they can identify individuals that may be more or less prone to a particular disorder (especially the disorders discussed herein) than individuals with the wild type gene. They can also be useful in research, in cell or tissue typing, in forensics, in diagnosis, etc.
  • a further aspect of the present invention is that of non-human animal, e.g. for use as an animal model, that has reduced expression of TREM-1 and/or of a TREM-1 ligand, relative to the wild type animal. Preferably it has no expression of TREM- 1 and/or of a TREM-1 ligand.
  • a further aspect of the invention is a non-human animal which also has reduced expression of TREM-3 relative to the wild type animal, and preferably has no expression of TREM-3.
  • Such animals are useful as a control compared to the wild type animal. They can be used to analyse the effectiveness of substances identified by the screening methods described earlier. They can also be used to assess side effects.
  • suitable animal models can be useful in reducing the overall number of test animals needed for screening for side effects, for drug efficacy, etc. Thus, these models can be beneficial in reducing overall animal suffering.
  • the non-human animal is a complete knock-out for the TREM-1 ligand or the TREM-1 receptor. Thus it does not produce functional TREM-1 ligand or functional TREM-1 receptor.
  • Breeding techniques can then be used to generate a line of mice that are homozygous for the modification.
  • transgenic animals may be provided have a plurality of genes knocked out relative to the wild type, especially TREM-3
  • TREM-1 TREM-3 double knock-out rodents particularly mice (TREM-1 -3 -/- mice) may be provided, as discussed later.
  • FIG. 1 shows the human and mouse TREM gene clusters.
  • TREM gene clusters are located on human chromosome 6 p.21.1 and mouse chromosome 17C. Both clusters include genes encoding Tremi, Trem2, Tremli (encodes TLT-1 ) and Treml2 (encodes TLT-2). Trem-1 and Trem-2 signal through the ITAM-containing adaptor DAP12.
  • TLT1 contains a cytoplasmic ITIM for recruitment of cytosolic phosphatases.
  • TLT-2 encodes a potential SH3 binding motif (+xPxxP, where + is arginine, x any amino acid, and P is proline).
  • the human TREM cluster also includes Ncr2, which encodes the NK cell receptor NKp44, while no murine NKp44 homolog has been identified. It is not yet known whether two additional human genes, Treml3 and Treml4, encode functional proteins.
  • the mouse TREM cluster includes genes encoding functional Trem-3 and Trem-L4 proteins. Trem3 is a pseudogene in human.
  • Figure 2 (lower plate )_shows canonical DAP12 signalling.
  • ITAM immunoreceptor tyrosine-based activation motif
  • crosslinking of receptors associated with DAP12 leads to phosphorylation of the tyrosines in the cytoplasmic ITAM motif by Src kinases. This leads to recruitment of SYK (or ZAP70) and subsequent phosphorylation of scaffolding molecules LAT and/or NTAL, and activation of PI-3K.
  • LAT/NTAL recruit several effectors: PLCy; TEC family members; the adaptor SLP76 in complex with Vav; the adaptor Grb2 in complex with Sos.
  • PI-3K produces Ptdlns(3,4,5)P3 (PIP3) which contributes to recruitment of PLCy , TEC, Vav to the cell membrane. All these intermediate signalling molecules lead to the recruitment/activation of Akt, c-CBL, and ERK, and to cytoskeletal remodelling (actin). PLCy generates the secondary messengers DAG and IP3 leading to activation of PKC ⁇ and calcium mobilization (Ca2+), respectively.
  • Figure 3 compares activating vs. inhibitory signalling of DAP12.
  • sepsis or simple endotoxemia with high LPS doses (right) lead to multivalent engagement of TREM-1 on neutrophils by TREM-1 ligand, generating a signalling cascade that synergises with that of TLR at different levels. This results in increased cytokine secretion and, possibly, cell adhesion and cell survival.
  • nonseptic conditions such as D-galactosamine-potentiated endotoxemia induce low occupancy of TREM-2 on macrophages by TREM-2 ligand, resulting in partial phosphorylation of DAP12 and recruitment of phosphatase SHP-1 or other inhibitory molecules that reduce cell responsiveness to TLRs.
  • FIG. 4 shows that DAP12 signalling augments mortality and inflammatory cytokine levels during endotoxemia.
  • A Survival of WT and DAP12-/- mice after endotoxemia was measured at three different doses, 5 mg/kg, 6.25 mg/kg, and 10 mg/kg. At both 5 and 6.25 mg/kg DAP12-/- mice had improved survival as compared to WT (p ⁇ 0.05 by Log-Rank Test). At 10 mg/kg both strains succumbed.
  • FIG. 5 shows that DAP12 signalling augments mortality and inflammatory events during bacterial sepsis WT and DAP12-/- mice were subjected to CLP and (A) survival and (B) cytokine production were assessed.
  • DAP12-/- mice are resistant to CLP as compared to WT (p ⁇ 0.001 by Log-Rank Test).
  • Plasma was harvested from WT or DAP12-/- mice 6 or 24 hours after CLP and cytokine levels were measured.
  • At 6 h we found equal levels of MCP-1 , IL-6 and TNF- ⁇ in WT and DAP12-/- mice.
  • WT mice had significantly higher levels of MCP-1 , IL-6, TNF- ⁇ and IL-10 (p ⁇ 0.05 by Mann-Whitney Test).
  • Figure 6 shows that DAP 12 signalling does not contribute to cellular recruitment or bacteriocidal activity. 24 hours after CLP, peritoneal exudate was harvested by peritoneal lavage. Total cell numbers (A), distribution of cell types (B) and bacterial load (C) were measured. We found no difference between WT and DAP12-/- mice.
  • FIG. 7 shows that DAP12 augments production by macrophages after sepsis but not sterile peritonitis.
  • WT and DAP12-/- mice were subjected to CLP and peritoneal cells harvested after 24 hours. Cells were cultured ex-vivo with or without stimulation with LPS (1 ⁇ g/ml) and levels of cytokine in the supernatant were measured. With no stimulation, WT cells (solid bars) produced more IL-6, MCP-1 , TNF- ⁇ and IL-10 as compared to DAP 12-/- cells (hashed bars) (p ⁇ 0.05 by Mann-Whitney Test). After LPS stimulation, WT cells produced increased amounts of TNF- ⁇ , MCP-1 , and IL-10.
  • Figure 8 shows ERK phosphorylation after stimulation of peritoneal exudates cells (PES) with LPS in vitro. 24 hrs after CLP, PEC were harvested, stimulated with LPS for various time points, and cell lysates were resolved by SDS-PAGE and immunoblotted for phospho-ERK1/2. Total ERK was determined as loading control. WT mice showed significant more phosphorylation than DAP12-/- mice after 30 minutes of stimulation.
  • PES peritoneal exudates cells
  • FIG. 9 illustrates the generation of TREM-1/TREM-3 deficient mice
  • FIG 10 shows staining results obtained with newly generated anti-TREM-3 antibodies.
  • HEK293 cells transfected with TREM-3 left or untransfected HEK293 cells were stained with either mAb 87.1 or mAb 12.7 (filled histograms) or control antibody (open histograms). Both antibodies specifically recognize the TREM-3 receptor.
  • FIG 11 shows flow cytometric results of bone marrow granulocytes of WT and TREM-1/-3 mice.
  • Whole bone marrow was stained with anti-CD1 1 b, -Ly6G-C (GR-1 ), -TRE M- 1 and -TREM-3 antibodies. Stained cells were analyzed by flow cytometry. All mice exhibit a similar population of CD1 1 b+/Ly6G-C+ granulocytes (left column).
  • WT granulocytes express TREM-1 and TREM-3 (middle and right columns, top panels) whereas TREM-1/3-/- do not .
  • Figure 12 shows survival of TREM-1/3-/- mice.
  • TREM-1/3+/+ and TREM-1/3-/- mice were subjected to a CLP sepsis challenge. Mice were subjected to a 2x #25 CLP injury and monitored for survival. TREM-1/3-/- mice were resistant to CLP as compared to wild type (WT).
  • Figure 13 shows % survival in respect of a murine model of pulmonary sepsis using a
  • FIG. 14 shows that TREM-1 ligand is expressed on neutrophils during sepsis or in vitro activation with PMA/ionomycin.
  • FIG. 14(A) shows a TREM-1 tetrameric construct.
  • the carboxy terminus of TREM-1 ectodomain is fused with a BirA tag and a 6-histidines tag. After biotinylation of the BirA sequence, TREM-1 monomers are assembled into fluorescent tetramers using PE-labeled streptavidin.
  • Figure 14(B) provides a FACS analysis of neutrophils purified from blood and stained with TREM- 1 tetramer and anti-CD16 antibody.
  • TREM-1 tetramers bind a subset of CD16+ neutrophils from a septic patient but not neutrophils from a healthy donor.
  • control tetramers CD69
  • CD69 control tetramers
  • Figure 14(C) shows a FACS analysis of neutrophils activated with PMA/I.
  • TREM-1 tetramers bind neutrophils of a healthy donor after treatment with PMA/I, whereas they do not bind unstimulated neutrophils. Control tetramers do not bind neutrophils stimulated with PMA/lonomycin.
  • Figure 15 shows a FACS analysis of the hTREM-1 ligand positive subpopulation of neutrophils isolated from septic patients. These cells were positive for CD1 1 b, CD10, CD66b, CD55 and CD35, all markers known to be expressed on circulating mature neutrophils. Neutrophils from a healthy donor also express these markers but do not bind hTREM-1 tetramer.
  • FIG 16 shows that monoclonal antibody R33 blocks binding of TREM-1 tetramers on septic neutrophils.
  • Neutrophils isolated from septic patients were preincubated with either R33 or an isotype matched control antibody (T2ctr).
  • Preincubation with R33 abrogates binding of TREM- 1 tetramers while the isotype matched control has no impact on tetramer binding.
  • mAb R33 recognises the TREM-1 ligand on the cell surface.
  • Figure 17 shows the results of screening a buffy coat cDNA library from sepsis patients for R33 antigen expression using the R33 monoclonal antibody.
  • Panel A FACS analysis of 293 cells transiently transfected with plasmids isolated from the human buffy coat cDNA library following FACS sorting of R33 positive cells. These cells are stained with R33 followed by goat anti rat Ig conjugated to PE.
  • Panel B Enrichment of R33 positive cells following 293 transfection with plasmids isolated from Plate F (149 colonies).
  • Panel C Further enrichment of R33 positive cells from Plate F.
  • Figure 18 shows a human CD177 amino acid sequence.
  • the signal peptide (amino acid 1-21 ) is shown in italics.
  • a GPI-anchor amidated glycine is shown in bold and is underlined (amino acid 408).
  • Figure 19 shows the mouse CD177 amino acid sequence.
  • the signal peptide (amino acid 1-21 ) is shown in italics.
  • the mouse sequence is approximately twice as long as the human sequence (excluding the leader sequence). This is likely have arisen due to a gene duplication event, because two parts of the sequence have a high degree of sequence identity with each other and with the human CD177 sequence, as is best illustrated in Figure 20.
  • Figure 20 shows an alignment between the human CD177 amino acid sequence and each of two parts of the mouse sequences. It can be seen that the human sequence has significant sequence identity with each part of the mouse sequence. This may indicate a gene duplication event in the mouse.
  • Figure 21 A shows a cDNA nucleotide sequence encoding the human CD177 amino acid sequence shown in Figure 18.
  • Figure 21 B shows a cDNA nucleotide sequence encoding the mouse CD177 amino acid sequence shown in Figure 19.
  • Figure 22 shows that a TREM-1 tetramer binds to septic patient neutrophils and not to resting neutrophils.
  • Figure 23 shows that the anti-mCD-177 antibody Y176 binds to neutrophils and a subset of monocytes in murine peripheral blood.
  • FIG. 24A shows TREM-1 ligand is specifically expressed on peripheral neutrophils of patients with sepsis.
  • the ratio between the geometric mean fluorescence of staining with TREM-1/lgM and human IgM is reported (GMF ratio).
  • Black squares represent patients at the time of admission into the ICU.
  • Black triangles represent the same patients at the time of clinical recovery.
  • White triangles represent patients with SIRS and no sign of infection.
  • Black diamonds represent healthy individuals.
  • Each data point represents GMF ratio of a single patient.
  • FIG. 24B shows that TREM-1 ligand expression is downregulated after recovery from sepsis.
  • Levels of expression of TREM-1 ligand were evaluated in patients with sepsis soon after admission into the ICU and at the time of clinical recovery. Data are expressed as the ratio between the geometric mean fluorescence (GMF) of cells stained with hTREM-1/lgM versus cells stained with control hlgM.
  • GMF geometric mean fluorescence
  • Figure 25 shows that R33 (anti-human CD177) blocks mTREM-1 binding to hCD177 transfected HEK293 cells.
  • Figure 26 shows that mouse CD177 is expressed on neutrophils and monocytes.
  • Figure 27 provides evidence of mTREM-1 binding to mCD177 (see discussion in Example 20).
  • the innate immune system In response to tissue damage or microbial products, the innate immune system initiates an inflammatory response tasked to eradicate the invading microbes 1"4 . In the context of disseminated infection or extensive tissue damage this immune response can become dysregulated, precipitating a systemic inflammatory response and a compensatory anti- inflammatory response 5"9 . The clinical consequence of this inappropriate immune activation is the sepsis syndrome, characterised by hypotension, organ failure and death 5 ' 7 ' 9 . Efforts to modulate the immune system during sepsis have met with limited success, and the "magic bullet" mediator of sepsis remains unidentified 10 ' 11 .
  • TREM-1 Triggering Receptor Expressed on Myeloid Cells 1
  • cytokines a molecule expressed on neutrophils and monocytes 12 .
  • blockade of TREM-1 attenuates inflammation and dramatically decreases mortality in a clinically relevant experimental model of sepsis 14 .
  • TREM-1 is not required to initiate a response to microbial products, but instead suggest a model in which ligation of TREM-1 by its ligand causes the amplification of the immune response, synergizing with the Toll-like receptors (TLRs) and the Nod like receptors (NLR) to cause exaggerated cytokine release 12 ' 15 ' 16 .
  • TLRs Toll-like receptors
  • NLR Nod like receptors
  • TREM-1 is the founding member of a family of receptors expressed in granulocytes (neutrophils), monocyte/macrophages, dendritic cells (DC), osteoclasts and microglia called triggering receptors expressed on myeloid cells (TREM) 12 ' 17"19 .
  • TREMs are transmembrane glycoproteins of the immunoglobulin (Ig) superfamily encoded by a gene cluster that maps to human chromosome 6p21 and mouse chromosome 17C3 in linkage with the MHC (Fig. 1 ) 12 ' 19 ' 20 .
  • the TREM gene cluster encodes both activating and inhibitory receptors.
  • TREM-1 , TREM-2 and TREM-3 receptors contain a single extracellular Ig-like domain of the V-type, a transmembrane region with a charged residue (lysine) and a short cytoplasmic tail (Fig. 1 ).
  • TREM-3 exists only as a pseudogene in humans (Fig. 1 ).
  • TREM-1 , TREM-2 and TREM-3 associate with the protein adaptor DAP12 for cell surface expression, signalling and function (Fig. 2).
  • the cytoplasmic domain of DAP12 contains an immunoreceptor tyrosine-based activation motif (ITAM), which functions as docking site for protein tyrosine kinases Syk and ZAP70 21"23 .
  • ITAM immunoreceptor tyrosine-based activation motif
  • the TREM cluster includes at least two other genes encoding the TREM-related proteins, called TLT- 1 and TLT-2.
  • TLT- 1 is expressed in platelets, contains an immunoreceptor tyrosine-based inhibitory motif (ITIM) in its cytoplasmic tail and recruits protein tyrosine phosphatases 24 ' 25 (Fig. 1 ).
  • ITIM immunoreceptor tyrosine-based inhibitory motif
  • TLT-2 is expressed in B cells and macrophages, contains a proline-rich region in its cytoplasmic tail and its function is unknown 26 (Fig. 1 ). Additional TREM-like genes and pseudogenes have been predicted by computational analysis of the TREM genomic region (Fig. 1 ). TREMs share limited homology with other Ig gene superfamily members.
  • the closest TREM relative is NKp44, an activating NK cell receptor encoded by a gene closely linked to the TREM genes 27 . More distant relatives of TREMs include the CMRF-35 family members 28"31 .
  • the receptor for polymeric Ig (plgR) 32 also shares homology with the extracellular region of the TREM family. However, none of the TREM receptors, CMRF35 or NKp44 bind Ig. In fact, the ligands for all these receptors are as yet unknown.
  • TREM-1 and TREM-2 are the most extensively characterised. TREM-2 is primarily expressed in preosteoclasts and microglia 12 . A genetic defect in TREM-2 results in a human disease, Nasu-Hakola disease (NHD), characterised by severe bone abnormalities and brain demyelination 33 . TREM-2 is also expressed in bone marrow derived macrophages, thioglycollate-elicited macrophages and alternatively activated macrophages 34 ' 35 and modulates their cytokine responses to microbial products 35 ' 36 . TREM-1 is expressed in granulocytes (neutrophils) and monocytes/macrophages. Preliminary studies in human suggest that TREM-1 activates these cells in vitro and contributes to systemic inflammatory responses and sepsis during microbial infections in v/Vo 13 ' 14 .
  • TREM-1 amplifies inflammation and contributes to pathogenesis of sepsis.
  • TREM-1 Human TREM-1 is expressed on blood neutrophils and a subset of monocytes. In normal tissues, TREM-1 is selectively expressed on alveolar macrophages. These are long-lived effector cells in the lung, specialized in recognition and clearance of pathogens, phagocytosis of apoptotic or damaged cells and removal of macromolecules. Furthermore, TREM-1 is expressed at high levels in neutrophilic infiltrates and epithelioid cells in human skin and lymph nodes infected by Gram positive and Gram negative bacteria as well as fungi 14 ' 37 . The tissue distribution of TREM-1 expression has suggested a role in inflammation.
  • TREM-1 a potent chemoattractant for neutrophils
  • MCP-1 Monocyte chemoattractant protein-1
  • MIP-1 ⁇ macrophage inflammatory protein 1 ⁇
  • TREM-1 triggering induces granulocyte release of myeloperoxidase but not phagocytosis.
  • TREM-1 and TLRs cooperate with each other in inducing inflammation.
  • Monocyte secretion of TNF- ⁇ and IL-1 ⁇ in response to LPS is markedly upregulated when TREM-1 mAbs are used as a co-stimulus, demonstrating the ability of TREM-1 to amplify inflammatory responses initiated by TLR 13 ' 15 .
  • LPS and other TLR ligands upregulate TREM-1 expression, potentiating its pro-inflammatory function 13 ' 15 .
  • TREM-1 As an amplifier of inflammation in vivo, we generated a recombinant mouse soluble TREM-1 fused with the Fc part of human IgGI (mTREM-1-Fc). This TREM-1 -Fc should compete with the endogenous TREM-1 for binding TREM-1 ligands, neutralizing the biological activities of endogenous TREM-1.
  • mTREM-1-Fc In an animal model of LPS-induced endotoxemia, blocking TREM-1 signalling with mTREM-1-Fc reduced hyper-responsiveness and death 14 .
  • septic shock including intraperitoneal injection of live E. coli and caecal ligation and puncture (CLP), blocking TREM-1 also protected mice against shock and death 14 .
  • transgenic mice overexpressing the TREM-1 signalling adaptor, DAP12 developed high numbers of blood neutrophils as well as massive macrophage infiltration in the lung and are highly susceptible to LPS-induced shock 38 .
  • This phenotype may be explained in part by constitutive activation of the TREM-1/DAP12- dependent pathway.
  • overexpression of DAP12 increased hepatic granulomatous inflammation elicited by zymosan A, while blockade of TREM-1 reduced granuloma formation 39 .
  • TREM-1 TREM-1-induced inflammatory responses by granulocytes and macrophages, particularly in response to microbial components, and implicate TREM-1 as a potential target for therapeutic intervention in human diseases caused by excessive inflammatory responses to infections, such as septic shock.
  • sTREM-1 A soluble form of TREM-1 (sTREM-1 ) has been identified in the serum of patients with sepsis 40 as well as in the serum of animals involved in an experimental model of septic shock 41 . Moreover, sTREM-1 was detected in the bronchioalveolar lavage (BAL) of patients with pneumonia 42 . There are two possible origins for sTREM-1. One possibility is that sTREM-1 is generated by proteolytic cleavage or membrane shedding of surface expressed TREM-1. Alternatively, sTREM-1 may be generated by de-novo translation of an TREM-1 mRNA splice variant which codes for a secreted form of TREM-1.
  • TREM-1 transcript which lacks exon 3 that encodes the transmembrane region, has been reported 43 ' 44 .
  • the physiological role of sTREM-1 is not fully understood. This molecule may scavenge the TREM-1 ligand that is not immediately bound to the surface displayed TREM-1 , thereby blunting immune responses and providing local control in the setting of inflammation 40 . Indeed, controlled release of soluble forms of multiple receptors critical to immunologic signalling and the inflammatory response has been described. These include a soluble form of the IL-1 receptor (IL-1 decoy RII) 45 ' 46 , TNF- ⁇ receptor 47"50 , and L-Selectin receptor 51 . Consistent with a modulatory function of sTREM-1 , a synthetic peptide mimicking a short highly conserved domain of sTREM-1 (LP17,
  • TDSRCVIGLYHPPLQVY attenuated cytokine production by human monocytes in vitro and protected septic animals from hyper-responsiveness and death in vivo 4 ⁇
  • This peptide was efficient not only in preventing sepsis but also in treating sepsis once the deleterious effects of proinflammatory cytokines is initiated.
  • TREM-1/DAP12 signalling promotes granulocytes and macrophage-inflammatory responses.
  • TREM-1 transmembrane region contains a charged residue (lysine) that allows association with the adapter DAP12 52 .
  • DAP12 contains cytoplasmic immunoreceptor tyrosine-based activation motifs (ITAM) (Fig. 2).
  • DAP12-associated receptor induces tyrosine phosphorylation of the ITAM by Src kinases.
  • the phosphorylated ITAM recruits the protein tyrosine kinases Syk and ZAP70, triggering phosphorylation of multiple adaptors such as LAT, NTAL, Slp76 in complex with Vav and Grb-2 in complex with Sos.
  • DAP12 also induces activation of phosphatidylinositol 3-kinase (PI3-K), phospholipase C ⁇ 1/2 (PLC ⁇ 1/2), TEC kinases, c-Cbl, and other downstream signalling mediators 21"23 .
  • PI3-K phosphatidylinositol 3-kinase
  • PLC ⁇ 1/2 phospholipase C ⁇ 1/2
  • TEC kinases c-Cbl
  • cytoplasmic mediators trigger intracellular Ca 2+ mobilization, rearrangement of the actin cytoskeleton, activation
  • TLRs signal through the adapters MyD88 and TRIF (Fig. 3).
  • MyD88 recruits IRAK4, IRAKI and TRAF6, initiating a signalling cascade ultimately leading to activation of NF-kB 4 .
  • TRIF recruits TBK-1 and IKK ⁇ , which mediate phosphorylation of IRF-3 and transcriptional activation of IFN- ⁇ 4 .
  • TLRs signal through Src tyrosine kinases via
  • DAP12-mediated signalling clearly potentiates TLR-mediated inflammatory responses in vitro and in vivo.
  • sustained ERK activation is essential for activation of the transcription complex AP-1 , particularly c-Fos 54 ' 55 .
  • DAP12-signalling induces sustained intracellular calcium mobilization, which activates Ca 2 7calmodulin-dependent phosphatase calcineurin 56 ' 57 .
  • Calcineurin dephosphorylates nuclear factor of activated T cell (NFAT) transcription factors, leading to their nuclear translocation 56 ' 57 .
  • NFAT nuclear factor of activated T cell
  • DAP12-mediated AP-1 and NFAT activation may synergize with NF-kB activation induced by TLR, resulting in enhanced transcriptional activation of genes encoding inflammatory mediators (Fig. 3).
  • Fig. 3 A similar model was recently demonstrated in osteoclasts, where DAP12 and other ITAM-containing adaptors generate Ca 2+ signals that allow Receptor Activator of NF-kB (RANK) to induce of NFATd (NFAT2), a key transcription factor for osteoclastogenesis 58 .
  • RANK Receptor Activator of NF-kB
  • NFAT2 NFAT2
  • ⁇ 1 and ⁇ 2 integrins expressed on the cell surface of granulocytes and macrophages also contribute to inflammation by mediating cell-cell interactions and adhesion to extracellular matrix proteins 59 ' 60 .
  • the adhesive function of integrins depends on intracellular signals generated by chemokine receptors that modify conformation and surface distribution of integrins 61 . It is possible that DAP12 also generates intracellular signals, such as Vav phosphorylation, that contribute to integrin activation (Fig. 3).
  • Proinflammatory responses of granulocytes and macrophages are elicited through additional receptors, such as those for IgG Fc, formyl-peptides, the inflammatory cytokines IL-1 and TNF, CD40L and other TNF-superfamily members.
  • additional receptors such as those for IgG Fc, formyl-peptides, the inflammatory cytokines IL-1 and TNF, CD40L and other TNF-superfamily members.
  • DAP12 is a transmembrane signalling adaptor associated with a family of activating immunoreceptors including not only the TREMs, but also SIRP- ⁇ 1 , CD200R, MDL-1 , KIRs, Ly49s, NKG2C/E, and others 62 . These receptors are expressed on the surface of granulocytes (neutrophils), macrophages, DC, osteoclasts, microglia and NK cells.
  • TREM-1 knockout mouse To validate the function of TREM-1 in sepsis in vivo it is necessary to develop a TREM-1 knockout mouse. However, in the case of the mouse, the TREM-1 gene is adjacent to a highly homologous gene, TREM-3, likely the result of a gene duplication event.
  • TREM-3 is separated from TREM-1 by only 4 kb, is expressed in mouse macrophages and is strongly upregulated in response to LPS 19 . Like TREM- 1 , TREM-3 promotes cell activation through DAP12 19 . It is reasonable to assume that, given their sequence homology and structural similarities, these two gene products have similar or overlapping functions in the mouse and may recognize the same ligand or closely related ligands. In contrast, in human, TREM-3 is a pseudogene, it is not expressed at the protein level and therefore there is no potential overlap between TREM-1 and TREM-3. Thus, to model the effect of blocking TREM-1 in humans, we have generated a TREM-1/TREM-3 double knockout (TREM- 1/3-/-) mouse. Our results show that TREM-1 /3-/- mice are more resistant to CLP than WT mice, indicating that the TREM-1 /3-DAP12 signalling complex exacerbates inflammation in the context of authentic sepsis.
  • DAP12-/- peritoneal exudate cells recovered from septic mice produce less cytokines than wild type PECs.
  • TREM-2 (as opposed to TREM-1/3) as the receptor mediating the inhibitory effects of DAP12 during the in-vitro stimulation of bone-marrow derived macrophages with low concentrations of LPS, and possibly also in D-galactosamine potentiated endotoxemia.
  • siRNA small interfering RNA
  • TREM-2 35 and a putative low affinity TREM-2 ligand 36 are expressed on the surface of macrophages, suggesting that TREM-2 could provide a tonic inhibitory signal to macrophages.
  • Inhibitory signalling may be mediated by incomplete DAP12 phosphorylation and consequential recruitment of protein tyrosine phosphatase SHP-1 , a major cytosolic mediator of inhibition 65 ' 66 .
  • DAP12 signalling contributes to inflammation and mortality from endotoxemia
  • DAP12-/- mice To determine if DAP12 contributed to the in-vivo response to endotoxin, we subjected WT and DAP12-/- mice to intraperitoneal injection of LPS and monitored them for survival. DAP12-/- mice tolerated doses of 5 mg/kg and 6.25 mg/kg of endotoxin, which resulted in 60-100% mortality in WT mice (Fig. 4A). However, DAP12-/- mice were not completely refractory to endotoxin, as they succumbed to a dose of 10 mg/kg (Fig. 4A). Thus, DAP12 signalling contributes to endotoxemia, although it is not required for the response to LPS.
  • Endotoxin causes shock by inducing macrophage production of TNF- ⁇ and other proinflammatory cytokines 67 .
  • DAP12 signalling exacerbated endotoxemia by increasing cytokine production
  • cytokine levels peak in 1-3 hours of endotoxemia 68 .
  • both WT and DAP12-/- mice had elevated circulating levels of TNF- ⁇ , IL-6, MCP-1 and IL-10.
  • WT animals had significantly higher levels of TNF- ⁇ and IL-10.
  • TNF- ⁇ and IL-10 were reduced in WT mice and the levels were equal to the DAP12-/- animals (Fig. 4B).
  • Sepsis is associated with high circulating cytokine levels that contribute to shock 69 .
  • cytokine levels in the serum of WT and DAP 12-/- mice 6 and 24 hours after CLP. At 6 hours, WT and DAP 12-/- mice had measurable serum levels of IL-6, MCP-1 and TNF- ⁇ , but there was no difference between the two groups. Between 6 and 24 hours, WT serum cytokine levels increased dramatically such that by 24 hours after the onset of sepsis the WT mice had significantly higher levels of IL-6, MCP-1 , TNF- ⁇ and IL-10 than did DAP12-/- mice (Fig. 5B). These data demonstrate that DAP12 signalling contributes to cytokine production during sepsis.
  • the proteins identified in this unbiased approach were previously described as acute phase reactants. Positive acute phase proteins (proteins known to increase in response to inflammation, i.e. apolipoprotein A-IV 71 , hemopexin 72 and complement component 3 72 ) accounted for 3/7 identified proteins. Negative acute phase proteins (those known to decrease with inflammation) accounted for 4/7 proteins (major urinary protein 73 , antithrombin III 74 , gelsolin 75 and MHC Q10 76 ). For every individual protein, the acute phase response was attenuated in DAP12-/- mice. This was manifest as lower levels of positive acute phase proteins and higher levels of negative acute phase proteins.
  • Positive acute phase proteins proteins known to increase in response to inflammation, i.e. apolipoprotein A-IV 71 , hemopexin 72 and complement component 3 72
  • Negative acute phase proteins (those known to decrease with inflammation) accounted for 4/7 proteins (major urinary protein 73 , antithrombin III 74 , gelsolin 75
  • DAP12 is not required for recruitment of cells or bacterial killing.
  • DAP12-signalling augments in-vitro cytokine production and ERK signalling of peritoneal exudates cells obtained from septic mice
  • DAP12 contributes to death from septic peritonitis by increasing inflammation.
  • DAP12 is not required to recruit cells to the peritoneum or to mediate an antimicrobial response.
  • DAP12 signalling exacerbates the inflammatory response by amplifying inflammatory cytokine production.
  • TREM-1/TREM-3 double-deficient mice are less susceptible to CLP than WT mice
  • CD200R 79"82 CD200R 79"82 , IREM2 83 , MDL-1 84 , and others 62 .
  • DAP12-/- mice cannot be used to pinpoint the specific functions of TREM-1 in vivo.
  • To address the function of TREM-1 in vivo is essential to generate a knockout model.
  • TREM-1 gene is adjacent to a very similar gene, TREM-3 19 , which is likely to encode a protein which may have overlapping function with TREM-1 and may recognize the same ligand or closely related ligands.
  • TREM-3 is a pseudogene.
  • the TREM-1 -3 targeting construct was designed to delete exons 3 and 4 encoding the transmembrane and cytoplasmic domains that are required for association of TREM-1 with DAP12, and exon 1 of TREM-3, encoding the leader sequence of TREM-3 (Fig. 9).
  • Chimeras transmitted the TREM-1 -3 mutation with the neomycin resistance gene deleted.
  • mice homozygous for the deletion was produced.
  • theTREM-1/3-/- mice were backcrossed onto the C57BI/6 background and then intercrossed when >70% of the genome was derived from the C57BL6 strain (as measured by SSLP typing).
  • Consideration of the genetic background of the mice is critical in that the ES cells in which the locus was targeted are derived from a 129/0Ia strain of mice and there is an uncharacterised defect in DAP12 signalling in 129 strains of mice 86 .
  • TREM-1/3 On sepsis survival, we compared the response of TREM- 1/3+/+ and TREM-1/3-/- mice (both from parallel breedings to give homogenous 70% C57BL/6 / 30% 1290Ia background) to a CLP sepsis challenge; survival was monitored for 14 days. Studies were limited to male mice to avoid the confounding effects of the estrous cycle on sepsis survival. We found that TREM-1/3-/- mice were resistant to CLP as compared to WT (Fig. 12).
  • CLP cecal ligation and puncture
  • a cDNA encoding human TREM-1 ectodomain was cloned into a bacterial expression vector Pet 28 (kindly provided by Daved Fremont, Washington University School of Medicine), which incorporates a BirA tag and a 6- histidines tag on the carboxy terminus of the protein of interest (Fig. 14).
  • the BirA sequence is efficiently biotinylated with recombinant biotin ligase.
  • the polyhistidine tag can be used to purify the recombinant protein by nickel sepharose chromatography.
  • This protein (TREM-1 ectodomain-BirA-6H) was purified from bacterial inclusion bodies, refolded and then isolated utilizing FPLC. Subsequently, the protein was biotinylated.
  • Biotinylated TREM-1 was then incubated with streptavidin coupled to phycoerythrin (PE).
  • streptavidin contains 4 distinct high affinity (10 "12 M) biotin-binding sites, biotinylated TREM-1 ectodomain and PE-streptavidin form PE-labeled tetramers.
  • the resultant molecule, hTREM-1 tetramer has four hTREM-1 ectodomains displayed on the central streptavidin molecule coupled to PE.
  • TREM-1 peripheral blood mononuclear cells
  • the subpopulation of neutrophils that bound hTREM-1 tetramer was characterised using mAbs against cell lineage markers. This subpopulation was CD56-, CD3-, CD19-, and CD16 h ⁇ gh consistent with a neutrophilic pattern of receptors. Further analysis revealed that this subpopulation of neutrophils was positive for CD11 b, CD10, CD66b, CD55, and CD1 1 c all markers known to be expressed on circulating mature neutrophils 96 (Fig. 15) .
  • CD35 complement receptor 1
  • CD35 receptor 1 CD35 receptor 1
  • CD16 levels are abnormally low in setting of inflammation and infection 98 .
  • CD16 (Fey receptor III) levels were decreased in the septic patients' neutrophils as compared to controls (Fig. 14).
  • the percentage of neutrophils positive for the TREM-1 ligand varied between patients from approximately 25% up to 90%.
  • neutrophils were treated with a variety of stimuli including fMLP, TNF- ⁇ , LPS, IL-1 , and PMA/lonomycin. These agents were chosen because they all stimulate human neutrophils causing some preferential degranulation of neutrophil granules.
  • Neutrophils have several unique types of granules, including specific, azurophilic, gelatinase, and secretory vesicles. Each type of granule contains characteristic proteins. Once the granule is exocytosed, the membrane of mobilized granule remains part of the plasma membrane, thus displaying molecules previously intracellular.” This is a mechanism for the neutrophil to display new receptors rapidly upon stimulation. By stimulating the cells with different compounds, we hoped to ascertain whether the ligand was synthesised de novo upon activation or preformed in granules.
  • anti-human neutrophil antibodies were generated. Rats were immunised with ligand positive neutrophils isolated from septic patients. After three rounds of immunisation, the rats were sacrificed and their spleens were fused with mouse SP2/0 mouse myeloma cells. The resulting hybridomas were screened for the production of antibodies that:
  • a mAb (lgG2a), designated R33 was identified. The antigen recognized by R33 was upregulated on neutrophils from septic patients and neutrophils pretreated with PMA/ionomycin.
  • RNA storage buffer was added and samples were frozen at -80 0 C. The total cellular RNA was then isolated using the RNAEASY kit from InVitrogen. The quality of the RNA was assessed by the Agilent bioanalyzer. Once adequate amounts of high quality RNA were purified, the samples were pooled and a custom nonamplified cDNA library was constructed by OpenBiosystems for our use.
  • Fig. 17 Two positives plates, F (Fig. 17, panel B) and H were identified. Plate F had approximately 149 individual colonies. These colonies were divided into 4 pools and the plasmid DNA was isolated. Following transfection of the DNA into 293 cells, another round of screening by FACS staining was performed. Through this process the R33 antigen was narrowed down eventually to a single colony (Fig. 17, panel C). This was found to express CD177, a molecule which is expressed on neutrophils and a subset of monocytes.
  • the amino acid sequence of this molecule is shown in Figure 18.
  • the molecule has a GPI anchor. It also has an extracellular portion that is involved in binding to the TREM-1 receptor.
  • the cDNA sequence is shown in Figure 21 A. GPI linked proteins are often shed from a cell surface and found as soluble proteins in plasma and serum. This has been shown to be the case for members of this protein family. For example Klippel et al (Blood, 100, No 7, 2441-2448 [2002]) report this phenomenon for PRV-1.
  • a soluble form of the ligand can be generated and is useful in order to perform binding assays on cells expressing TREM-1. This can be achieved using a construct in our laboratory which encodes a mutated form (does not bind to Fc receptors) of the Fc portion of IgG.
  • the TREM-1 ligand gene can be fused in frame with the Fc.
  • This plasmid can be transfected into mammalian cells.
  • the protein product will be secreted into the media forming dimers via the Fc interaction.
  • the resulting protein product will be a Fc fusion protein with two ligand heads.
  • Supernatant from cells secreting this molecule can be collected and the molecule can be purified using a protein G column.
  • This construct is useful for assessing the binding of receptor and ligand in both directions, i.e. soluble TREM-1 binds surface expressed R33 antigen and soluble R33 antigen binds surface expressed TREM-1.
  • Our laboratory has used this strategy to characterize several ligand receptor interactions in the past 13 ' 14 ' 114 .
  • the soluble TREM-1 ligand Fc protein can then be incubated with both cells expressing TREM-1 naturally (neutrophils and monocytes) as well as cells transfected with TREM-1 encoding plasmids. Binding of the Fc fusion protein can be detected using an anti human Fc conjugated to PE and FACS analysis. The converse experiment can also be performed in which the ability of the human TREM-1 tetramer to bind a cell line transfected with the TREM-1 ligand can be assessed.
  • the TREM-1 ligand can be amplified from the cDNA library plasmid and subcloned into pcDNA3 vector. This vector contains a neomycin selection allowing for the production of stable mammalian transfectants.
  • the plasmid can be transfected into 293 cells and placed under antibiotic selection. Resistant cells can then be analyzed in FACS for staining with the R33 antibody. High expressing stable transfectants can be cloned and then used in binding assays with the TREM-1 tetramer molecule as well as the TREM-1 Fc molecule previously made in our laboratory.
  • a T cell hybridoma reporter cell line has been constructed which expresses the TREM-1 molecule fused to the cytoplasmic region of CD3 ⁇ . If the TREM-1 molecule is engaged in a functional way, ZAP70 is recruited to the CD3 ⁇ and a series of intracellular phosphorylation events lead to activation of PLCg and increased intracellular calcium.
  • the reporter cell contains a plasmid encoding the NFAT promoter fused to a sequence encoding green fluorescent protein (GFP). NFAT is activated by intracellular calcium mobilization. This allows one to co-incubate the TREM- 1 expressing GFP reporter with a putative ligand and then analyze the cells for GFP expression by FACS. Our laboratory and others have used this system to ascertain the biological relevance of other receptor ligand interactions 115 . This functional assay system can be used as another measure of the biological relevance of the TREM-1/TREM-1 ligand interaction.
  • the soluble TREM-1 ligand molecule as well as an irrelevant control Fc fusion protein can be incubated with freshly isolated human neutrophils and monocytes.
  • IL-1 , IL-8 and MPO activity will be measured in the neutrophils as surrogate markers of inflammation.
  • the effect of R33 antigen binding on neutrophil phagocytosis will also be assessed.
  • the secretion of TNF ⁇ , IL-8, and MCP-1 can be measured to ascertain the effects of TREM-1 engagement on these molecules. We expect engagement to result in the secretion of these proinflammatory cytokines.
  • Wild type and TREM1/3 deficient mice can be used to assess the impact of soluble TREM-1 ligand on murine sepsis. Survival, serum cytokine production, peritoneal infiltration and local and systemic bacterial load can be assessed in these mice. We predict that excess TREM-1 ligand in the knockout mice should have no impact on survival whereas excess TREM-1 ligand, if stimulatory should increase cytokine production and mortality in the wild type mice.
  • TREM-1 ligand we expect binding of the TREM-1 ligand to trigger proinflammatory cytokine production. In vivo we expect that administration of soluble TREM-1 ligand will result in increased cytokine production and increased mortality in wild type mice following CLP while the knockout mice should be unaffected by this molecule.
  • TREM-1 ligand as a marker of sepsis
  • TREM-1 ligand was assessed for the relationship between levels of expression of TREM-1 ligand and the clinical status of sepsis patients.
  • the levels of expression of TREM-1 ligand decreased at the time of discharge from the ICU (Fig. 24B).
  • the second determination of TREM-1 ligand could not be performed because the patient died from septic shock.
  • TREM- 1 ligand expression could not be detected at the time of admission into the ICU, despite documented systemic bacterial infection. This might have been due to the fact that inadequate blood samples with high cell mortality were delivered to the laboratory.
  • TREM-1 ligand expression is exclusively detected on peripheral neutrophils from patients with sepsis but not with SIRS of non-microbial origin, therefore representing a useful marker of sepsis.
  • Measurement of plasma levels of soluble TREM-1 has also shown its diagnostic accuracy in distinguishing sepsis from SIRS [Gibot S, Kolopp-Sarda MN, Bene MC, et al. Ann Intern Med 2004;141 :9-15].
  • advances in sepsis research require better markers than the ones available to delineate more homogenous subsets of patients within a highly heterogeneous group of critically ill patients, and to identify patients having the particular biological abnormality that a proposed therapy will target.
  • TREM-1 ligand might represent a useful diagnostic marker to predict the presence and severity of sepsis providing information in establishing a diagnosis to identify a patient who has the disease and therefore might respond to a particular therapy; quantifying the severity of sepsis to identify patients who are more likely to experience a beneficial outcome; measuring the response to therapy to determine how a patient is responding to an intervention.
  • TREM-1 ligand is an important mediator in sepsis and it is specifically expressed in patients with sepsis. Since intervention must not only be targeted to TREM-1 but it must be given at the appropriate time, the analysis of the expression of TREM-1 ligand during the evolution of the inflammatory response during sepsis is of fundamental importance in effective therapies for sepsis.
  • 293 cells alone or 293 cells transfected with murine CD177 were preincubated with different concentrations of test compounds for 30 minutes on ice and then incubated with soluble murine TREM-1 molecule (100ug/ml) for 45 minutes on ice, cells were then washed with FACS buffer (PBS, 2% BCS), incubated with anti human FC biotin for 20 minutes on ice, washed once with FACS buffer, and incubated with streptavidin APC for 20 minutes, following a wash with FACS buffer, the cells were immediately analyzed. Dead cells were excluded. A shift of the histogram to the left indicates that the test compound is inhibiting binding of the TREM-1 molecule to CD177.
  • FACS buffer PBS, 2% BCS
  • a T cell hybridoma reporter cell line has been constructed which expresses the TREM-1 molecule fused to the cytoplasmic region of CD3 ⁇ . If the TREM-1 molecule is engaged in a functional way, ZAP70 is recruited to the CD3 ⁇ and a series of intracellular phosphorylation events lead to activation of PLCg and increased intracellular calcium.
  • the reporter cell contains a plasmid encoding the NFAT promoter fused to a sequence encoding green fluorescent protein (GFP). NFAT is activated by intracellular calcium mobilization.
  • GFP green fluorescent protein
  • TREM- 1 expressing GFP reporter This allows one to co-incubate the TREM- 1 expressing GFP reporter with CD177 - either in a soluble form or expressed by a transfected cell line - in the presence or absence of different concentrations of test compounds and then analyze the cells for GFP expression by FACS. Inhibition of activation of the TREM-1 reporter cell line by CD177 indicates that the test compound binds to TREM-1.
  • the above system can be modified by using different reporter systems, such as lacZ.
  • a TREM-1 ligand e.g. CD177
  • a TREM-1 ligand binding portion thereof can be obtained in pure form.
  • This can then be inoculated into an animal and used to generate a series of hybridomas producing monoclonal antibodies.
  • the antibodies can then be screened using the screening procedures of the present invention in order to identify ones that block or reduce the binding of the TREM-1 ligand to a TREM-1 receptor.
  • Such antibodies can then be used in diagnostic tests to diagnose sepsis (especially of microbial origin, e.g. of bacterial or fungal origin).
  • a control may be used based upon neutrophils or monocytes from a healthy patient.
  • This test can also distinguish between sepsis of microbial origin and non-microbial derived SIRS. In the latter case (unlike the former) there is no substantial binding of the antibodies to the neutrophils or monocytes obtained from a patient.
  • Monoclonal antibodies to the TREM-1 ligand can be raised and screened as discussed in Examples 13-14.
  • Anitibodies identified by screening as being successful in blocking the binding of TREM-1 to its receptor can then be used for further testing.
  • Antibodies that are successful in this test can be selected for further analysis, including safety testing and possible eventual clinical trials.
  • Clinical trials can be performed by comparing the results of the antibodies on a patient group with microbial sepsis with results for a patient group of non-microbial origin.
  • the trails will be successful if there are no major side effects with either group and there is a significant improvement in the condition of the patient group with microbial sepsis, relative to the patient group with SIRS of non-microbial origin.
  • Appropriate control groups can also be used, e.g. patients with microbial sepsis who are given a placebo, patients with SIRS of non-microbial origin who are given a placebo, a group of healthy volunteers that are given a placebo and a group of healthy volunteers that are given the antibody.
  • the antibodies can be provided in a form that reduces cross-reactivity.
  • they can be “humanised” or even “completely human”, as discussed earlier.
  • They can be provided in a sterile pharmaceutical composition together with one or more substances that help extend the half life in vivo (e.g. pegylation can be used as discussed earlier). They can be administered by any appropriate route, but are preferably provided as an injectable composition.
  • Dosage ranges are given earlier, but can of course be optimised by the results of animal trials before administration to humans. If side effects develop at a certain dosage then the dosage should of course be reduced as appropriate.
  • CD177 and of other TREM-1 ligands e.g. PRV-1
  • Antibodies to different variants can be useful in purification, diagnosis, treatments, tissue typing, comparative studies, assessments of specificity, etc.
  • This example illustrates the generation of antibodies to murine CD177.
  • Murine CD177 was identified using blast homology searches. Specific primers were generated and used to amplify the CD177 sequence from murine cDNA. The resulting fragment was subcloned into the expression vector pCDNA ⁇ . This plasmid was transfected into 293 cells and stable high expressing cells were isolated using high efficiency cell sorter. These cells were then utilized to immunize rats. Subsequently the rat lymph node was fused to SP2/0 cells and following HAT selection, individual antibody producing clones were isolated and screened for binding to the recombinant mouse CD177 molecule. Forty positive clones were identified. One of these antibodies was then purified and biotinylated (Y176) to be used to ascertain where CD177 was expressed in the mouse.
  • biotinylated Y176
  • Bone marrow was harvested and incubated with FcBlocking supernate. Following a 20 minute room temperature incubation, biotinylated Y176 (followed by streptavidin APC), anti CD11 b fitc and anti GR1 PE was used to characterize the CD177 positive population. Examination of bone marrow revealed CD177 is expressed on inflammatory monocytes and neutrophils.
  • R33 (anti-human CD177) blocks mTREM-1 binding to hCD177 transfected HEK293 cells
  • HEK293 cells transfected with the human CD177 full length were analyzed by cytofluori metric analysis, as shown in Figure 25.
  • the grey histogram represents staining with soluble mouse TREM1/lgG in the presence of an isotype control MAb.
  • the dashed histogram represents staining with soluble mouse TREM1/lgG in the presence of the R33 MAb. Staining with a control soluble mouse TLT/lgG is represented by the white histogram.
  • the data show that the the R33 MAb specifically blocks binding of soluble mouse TREM1/lgG to CD177-transfected cells.
  • Mouse CD177 is expressed on neutrophils and monocytes
  • LEFT The dot plot represents forwards vs size scatter of mononuclear cells in the mouse peripheral blood. Based on physical parameters, three gates were constructed that identify different subsets: a) lymphocytes, b) monocytes, c) neutrophils.
  • Murine TREM-1 soluble molecule binds to 293 cells transfected with murine CD177
  • 293 cells alone or 293 cells transfected with murine CD177 were incubated with soluble murine TREM-1 molecule (100ug/ml) for 45 minutes on ice, cells were then washed with FACS buffer (PBS, 2% BCS), incubated with anti human FC biotin for 20 minutes on ice, washed once with FACS buffer, and incubated with streptavidin APC for 20 minutes, following a wash with FACS buffer, the cells were immediately analyzed. Dead cells were excluded.
  • FACS buffer PBS, 2% BCS
  • Beutler B Inferences, questions and possibilities in Toll-like receptor signalling. Nature. 2004;430:257-263.
  • Hotchkiss RS Karl IE. The pathophysiology and treatment of sepsis. N Engl J Med. 2003:348:138-150.
  • TERT-1 Triggering receptor expressed on myeloid cells-1 amplifies the signals induced by the NACHT- LRR (NLR) pattern recognition receptors. J Leukoc Biol. 2006.
  • TREM-3 an activating receptor on mouse macrophages: definition of a family of single Ig domain receptors on mouse chromosome 17. Eur J Immunol. 2002;32:59-66.
  • the human TREM gene cluster at 6p21.1 encodes both activating and inhibitory single IgV domain receptors and includes NKp44. Eur J Immunol. 2003;33:567-577.
  • McVicar DW Burshtyn DN. Intracellular signalling by the killer immunoglobulin-like receptors and Ly49. Sci STKE. 2001 ;2001 :RE1.
  • TREM family member, TLT-1 is found exclusively in the alpha-granules of megakaryocytes and platelets. Blood. 2004;104:1042-1047.
  • TREM-like transcript (TLT)-I a putative inhibitory receptor within the TREM cluster. Blood. 2002; 100:3822- 3824.
  • Trem-like transcript 2 is expressed on cells of the myeloid/granuloid and B lymphoid lineage and is up-regulated in response to inflammation. J Immunol. 2006;176:6012-6021.
  • the CMRF-35 mAb recognizes a second leukocyte membrane molecule with a domain similar to the poly Ig receptor, lnt Immunol.
  • CMRF-35-like molecule-1 a novel mouse myeloid receptor, can inhibit osteoclast formation. J Immunol. 2003;171 :6541-6548.
  • Humphrey MB Daws MR, Spusta SC, et al. TREM2, a DAP12-associated receptor, regulates osteoclast differentiation and function. J Bone Miner Res. 2006;21 :237-245.
  • Mounzer KC Moncure M
  • Smith YR Dinubile MJ. Relationship of admission plasma gelsolin levels to clinical outcomes in patients after major trauma. Am J Respir Crit Care Med. 1999;160:1673-1681.
  • CD200 is a ligand for all members of the CD200R family of immunoregulatory molecules. J Immunol. 2004;172:7744-7749.
  • Myeloid DAP12-associating lectin (MDL)-I is a cell surface receptor involved in the activation of myeloid cells. Proc Natl Acad Sci U S A. 1999;96:9792-9796.
  • Hotchkiss RS Dunne WM, Swanson PE, et al. Role of apoptosis in Pseudomonas aeruginosa pneumonia. Science. 2001 ;294:1783.
  • Clark RA Nauseef WM. Preparation and functional analysis of other human lymphoid and nonlymphoid cells. In: Coligan JE, Kruisbeek AM, Margulies DH, Shevach EM, Strober W, eds. Current Protocols in Immunology. Vol. 2: John Wiley & Sons, Inc.; 1999:7.23.21-27.23.17.
  • Ig-like transcript 2 (ILT2)/leukocyte Ig-like receptor 1 (LIR1 ) inhibits TCR signalling and actin cytoskeleton reorganization. J Immunol. 2001 ;166:2514-2521.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • Molecular Biology (AREA)
  • Urology & Nephrology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Biotechnology (AREA)
  • Food Science & Technology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • Pathology (AREA)
  • General Physics & Mathematics (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Public Health (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Virology (AREA)
  • Oncology (AREA)
  • Peptides Or Proteins (AREA)
  • Communicable Diseases (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

Un ligand de TREM-1 est identifié. Ceci permet de fournir/identifier divers dérivés qui sont capables de se lier au récepteur de TREM-1. Le ligand de TREM-1 ou les dérivés peuvent être utilisés dans le criblage de médicaments/candidats médicaments. Les substances qui bloquent ou réduisent la fixation de ligand de TREM-1/dérivé à un récepteur de TREM-1 peut être utile pour traiter une septicémie, en particulier une septicémie d'origine bactérienne ou fongique. Les anticorps contre le ligand peuvent être utiles dans le diagnostic d'une septicémie, en particulier une septicémie d'origine bactérienne ou fongique.
EP08786360A 2007-07-23 2008-07-23 Criblage, thérapie et diagnostic Withdrawn EP2174131A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US96161507P 2007-07-23 2007-07-23
PCT/EP2008/059668 WO2009013319A1 (fr) 2007-07-23 2008-07-23 Criblage, thérapie et diagnostic

Publications (1)

Publication Number Publication Date
EP2174131A1 true EP2174131A1 (fr) 2010-04-14

Family

ID=39816994

Family Applications (1)

Application Number Title Priority Date Filing Date
EP08786360A Withdrawn EP2174131A1 (fr) 2007-07-23 2008-07-23 Criblage, thérapie et diagnostic

Country Status (5)

Country Link
US (1) US20100306863A1 (fr)
EP (1) EP2174131A1 (fr)
JP (1) JP2010534828A (fr)
CN (1) CN101821621A (fr)
WO (1) WO2009013319A1 (fr)

Families Citing this family (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8393201B2 (en) 2010-09-21 2013-03-12 Webtech Wireless Inc. Sensing ignition by voltage monitoring
RU2463606C1 (ru) * 2011-08-17 2012-10-10 Учреждение Российской академии медицинских наук Научно-исследовательский институт комплексных проблем сердечно-сосудистых заболеваний Сибирского отделения Российской академии медицинских наук (УРАМН НИИ КПССЗ СО РАМН) Способ ранней диагностики осложненного системного воспалительного ответа у пациентов, оперированных в условиях искусственного кровообращения
CN102517287B (zh) * 2012-01-10 2013-09-11 广西大学 抑制水牛TREM1基因表达的shRNA、慢病毒表达载体及其构建方法和应用
RS57827B1 (sr) 2012-02-15 2018-12-31 Novo Nordisk As Antitela koja vezuju protein 1 prepoznavanja peptidoglikana
JP6307080B2 (ja) 2012-09-07 2018-04-04 インセルム(インスティチュート ナショナル デ ラ サンテ エ デ ラ リシェルシェ メディカル) 骨髄細胞に発現する誘発性受容体1(trem−1)trem様転写産物1(tlt−1)に由来する阻害ペプチドおよびその使用
EP2835641A1 (fr) * 2013-08-09 2015-02-11 Inotrem Procédés et kits pour prédire le risque de survenue d'un accident cardiovasculaire chez un sujet
JP6752221B2 (ja) * 2015-04-13 2020-09-09 リジェネロン・ファーマシューティカルズ・インコーポレイテッド ヒト化sirpa−il15ノックインマウス及びその使用方法
CA3091047A1 (fr) * 2018-02-14 2019-08-22 Yale University Compositions pour la modulation d'une proteine trem ou treml et procedes d'utilisation
US20220153806A1 (en) * 2019-03-01 2022-05-19 University Of Tsukuba Composition for use in treatment of allergic diseases
WO2021011678A1 (fr) * 2019-07-15 2021-01-21 Bristol-Myers Squibb Company Anticorps anti-trem-1 et leurs utilisations
CN111157741B (zh) * 2019-12-30 2022-08-16 广州市妇女儿童医疗中心 髓系细胞触发受体1在制备胃炎诊断或治疗试剂中的应用及试剂盒
CN112526143B (zh) * 2020-12-03 2021-08-03 中山大学附属第五医院 髓系细胞触发受体2作为新型冠状病毒肺炎诊断或治疗靶点的应用
CN114736957B (zh) * 2022-04-15 2024-03-12 中国人民解放军空军军医大学 外泌体介导rna casc15干预阿尔茨海默症突触损失的应用
CN116908446B (zh) * 2023-06-27 2024-05-07 广州市妇女儿童医疗中心 用于诊断川崎病冠状动脉瘤的生物标志物及其应用

Family Cites Families (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4444887A (en) * 1979-12-10 1984-04-24 Sloan-Kettering Institute Process for making human antibody producing B-lymphocytes
US4522811A (en) * 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
US4716111A (en) * 1982-08-11 1987-12-29 Trustees Of Boston University Process for producing human antibodies
IT1164225B (it) * 1983-05-13 1987-04-08 Anic Spa Analoghi retro-invertiti del pentapeptide potenziante la bradichina bpp5a e metodi per la loro preparazione
US5413923A (en) * 1989-07-25 1995-05-09 Cell Genesys, Inc. Homologous recombination for universal donor cells and chimeric mammalian hosts
EP1690935A3 (fr) * 1990-01-12 2008-07-30 Abgenix, Inc. Génération d'anticorps xenogéniques
US5625126A (en) * 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5661016A (en) * 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5814318A (en) * 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
US5633425A (en) * 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5545806A (en) * 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
DK0546073T3 (da) * 1990-08-29 1998-02-02 Genpharm Int Frembringelse og anvendelse af transgene, ikke-humane dyr, der er i stand til at danne heterologe antistoffer
WO1993011236A1 (fr) * 1991-12-02 1993-06-10 Medical Research Council Production d'anticorps anti-auto-antigenes a partir de repertoires de segments d'anticorps affiches sur phage
US5916771A (en) * 1996-10-11 1999-06-29 Abgenix, Inc. Production of a multimeric protein by cell fusion method
US20090081199A1 (en) * 2001-03-20 2009-03-26 Bioxell S.P.A. Novel receptor trem (triggering receptor expressed on myeloid cells) and uses thereof
CA2342376C (fr) * 2001-03-20 2013-11-12 Marco Colonna Nouveau recepteur trem (recepteur activateur exprime dans les cellules myeloides) et ses utilisations
GB0305478D0 (en) * 2003-03-10 2003-04-16 Bioxell Spa Diagnostic and prognostic compounds and method

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2009013319A1 *

Also Published As

Publication number Publication date
US20100306863A1 (en) 2010-12-02
WO2009013319A1 (fr) 2009-01-29
JP2010534828A (ja) 2010-11-11
CN101821621A (zh) 2010-09-01

Similar Documents

Publication Publication Date Title
US20100306863A1 (en) Screening, therapy and diagnosis
US20200254058A1 (en) Therapeutic trem-1 peptides
Lai et al. Immune sensing of cell death through recognition of histone sequences by C-type lectin-receptor-2d causes inflammation and tissue injury
US20090169543A1 (en) Method for regulating immune function in primates using the Foxp3 protein
JP2008522162A (ja) Merの診断用および治療用の作用薬
Brouns et al. The structure of the μ/pseudo light chain complex on human pre‐B cells is consistent with a function in signal transduction
WO2010132370A2 (fr) Peptides solubles de la famille trem-1 et procédés d'utilisation
O'Shea et al. Exit of the pre-TCR from the ER/cis-Golgi is necessary for signaling differentiation, proliferation, and allelic exclusion in immature thymocytes
CN113840618A (zh) 一种通过调节abcf1进行免疫调节的方法
US7534571B2 (en) Diagnostic and prognostic compounds and method
AU2005234705B2 (en) Therapeutic peptides and method
US20060008891A1 (en) Cathepsin G-related peptides as modulators of formylpeptide receptors (FPR)
Stappers et al. Recognition of DHN-melanin by MelLec, is required for protective immunity to Aspergillus
US20190263906A1 (en) Immune modulators for reducing immune-resistance in melanoma and other proliferative diseases
KR101268562B1 (ko) Tlt-6 단백질에 대한 항체 및 그 응용
Meyermann Investigations on the C-type lectin receptor Clec12a and its role in the activation by the adjuvant alum
Meyermann TUM School of Medicine and Health
Finnegan An investigation into the role of Pellino2 in lung inflammation
US20070099252A1 (en) Diagnostic application of mail
Corlianò LPS-dependent NFAT activation in dendritic cells is regulated by IP4-mediated calcium entry through plasma membrane IP3R3
Müller Role of Syntaxin-11 and Munc18-2 in lymphocyte cytotoxic granule exocytosis
Ramiscal An analysis of the putative role of ROQUIN as a T cell RING-directed ubiquitin ligase
Perretti et al. Workshop 1: Emerging aspects of phagocyte biology
BAUMANN et al. R. 1 FcyRI, FcyRIII and CSaR are co-dominant in the initiation of the inflam-matory cascade
Chen et al. Broad and direct interaction between TLR and Siglec families of pattern recognition

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20100120

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA MK RS

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: NOVO NORDISK A/S

17Q First examination report despatched

Effective date: 20100608

DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN WITHDRAWN

18W Application withdrawn

Effective date: 20120320